Skip to main content

Full text of "USPTO Patents Application 10822231"

See other formats


(12) INTERNATIONAL APPLICAtlC>y^UBLISHED UNDER THE PATENT COOPERATION TREATY (PCT) 

(19) World Intellectual Property 
Organization 
Internationa] Bureau 



(43) International Publication Date 
23 June 2005 (23.06.2005) 




PCT 



(10) International Publication Number 

WO 2005/056759 A2 



(51) International Patent Classification 7 : 



C12N 



(21) International Application Number: 



PCTAJS2004/040694 



(22) International Filing Date: 3 December 2004 (03.12.2004) 



(25) Filing Language: 

(26) Publication Language: 



English 
English 



(30) Priority Data: 

60/527,167 
60/581,613 
60/601,665 
60/619,483 



4 December 2003 (04. 12.2003) US 

21 June 2004 (21.06.2004) US 

13 August 2004 (13.08.2004) US 

1 6 October 2004 (16.1 0.2004) US 



(71) Applicant (for all designated States except US): XEN- 
COR,INC. [US/US]; 111 W. Lemon Avenue, Monrovia, 
CA 91016 (US). 

(72) Inventors; and 

(75) Inventors/Applicants (for US only): LAZAR, Gregory, 
Alan [US/US]; Apt. C, 839 E. Mobeck Street, West Cov- 
ina, CA 91790 (US). DESJARLAIS, John R. [US/US]; 



2096 E. Crary Street, Pasadena, CA 91104 (US). HAM- 
MOND, Philip W. [US/US]; 421 Crestvale Drive, Sierra 
Madre, CA 91024 (US). 

(74) Agents: BOOTH, Paul, M. et ah; Heller Ehrman White & 
McAuliffe LLP, Suite 300, 1666 K Street, NW, Washing- 
ton, DC 20006- 1 228 (US). 

(81) Designated States (unless otherwise indicated, for every 
kind of national protection available): AE, AG, AL, AM, 
AT, AU, AZ, B A, BB, BG, BR, BW, BY, BZ, CA, CH, CN, 
CO, CR, CU, CZ, DE, DK, DM, DZ, EC, EE, EG, ES, FI, 
GB, GD, GE, GH, GM, HR, HU, ID, 1L, IN, IS, JP, KE, 
KG, KP, KR, KZ, LC, LK, LR, LS, LT, LU, LV, MA, MD, 
MG, MK, MN, MW, MX, MZ, NA, Nl, NO, NZ, OM, PG, 
PH, PL, PT, RO, RU, SC, SD, SE, SG, SK, SL, SY, TJ, TM, 
TN, TR, TT, TZ, UA, UG, US, UZ, VC, VN, YU, ZA, ZM, 
ZW. 

(84) Designated States (unless otherwise indicated, for every 
kind of regional protection available): ARIPO (BW, GH, 
GM, KE, LS, MW, MZ, NA, SD, SL, SZ, TZ, UG, ZM, 
ZW), Eurasian (AM, AZ, BY, KG, KZ, MD, RU, TJ, TM), 
European (AT, BE, BG, CH, CY, CZ, DE, DK, EE, ES, FI, 
FR, GB, GR, HU, IE, IS, IT, LT, LU, MC, NL, PL, PT, RO, 

[Continued on next page] 



(54) Title: METHODS OF GENERATING VARIANT PROTEINS WITH INCREASED HOST STRING CONTENT AND COM- 
POSITIONS THEREOF 



< 
On 

IT) 
IT) 

o 



Kahat 

VH 1-2 
VlTl-S 
VH~l-8 
VH_2-18 
VH 1-24 
VH~l-45 
VH 1-46 
VH_1-SB 
VH~l-6» 
VH_l-f 

VH 2-5 

VH>-26 

VH~2-70 

VH3-7 
VH 3-9 
' VH_3-11 
VH 3-13 
VH 3-15 

VH3-16 
VHJJ-19 
VH_3-20 
VH_3-21 
VH_3-23 
VH_3-30 
VH_3-30-3 
VH 3-33 
VH>-35 
VH_3-3S 
VH3-43 
VH_3-4B 
VH 3-4S 
VH_3-53 
VH__3-64 
VH3-S6 
VH 3-72 
VH3-73 



Human Germ line VH Sequences 

CD* 1 FM CDB3 FE3 ctSU 

1 2 3 4 5 6 7 6 9 in 

1 ««SCT$9Q1234S67890X234S67e 901^^789012*5^^^ 

Q^kVQSGAgVKKraASVKVSCKASOYT. FreTDIj<_WYKQftTQQGl£K>tC ?»e<P RSSmi TAQ^gGH^TKTWrS I STATKHLSSLRSEryTAVYYCAS 
^^^^^^ S ^^ CKftSGf 2^ rG ^ KVQ ^^KGCKI^WICMIW G6GMTWYAQgPtIigH VTITgIlKgTgrAYMgT-Qgt Denrvri^pvTri ^ 
0fVn^E5Cn>ALVICPTQTXTL7XTFSOFSI^T^ajgy5W I RQPPCXXUWI AL ID KDDOXYrST^FjCTgT/rr SXOTKTOOVVLra K^^V^l ATTTCARI 



gV«iV^3OT^VQroQSIJtLSCAAaOPTF 8STVW8 WVBQftPGKGUWV Attl KQ 



" I JTL : ~ ^^w. . , ■■. n,*^^ i^.t,. i ^ t. anv jVj^TTISEOSJJCBSLTl^KHSIJUKin'AVyYCAR 

EVQLVESqCXn^VQPCaglJ.LaaUl^FTT^rAMg WVRCSATCM^rvS^ HBGSICTrMWV^B msimi^^.^^ci P >^^!^ 

^LVHSa^^POC^I^UVSGrrF 3 gTt^_HV^ aoitt ^55v^5 rftt eraOU^TLS^^^AVYTSS 

HVQLVESGGGLVia^SSlja^CAASGITrSlffiWffi WVRQAJCgElJWV ra ^ i ^ ^ 

BVC^VE5gX3iVgPGGSLaiSCAASGFTFSNSEMN HARKftrQ3SL5WY 3(3YBH HQSRTHYVDSVKR RFI I SimBSrarebYI^RRKAEatAVyTUVH 
TVOLV-ESGOGLVSPO^ J?I .SCAASCnTSHSraM W VRQM€ZGLg#g 8CVCT HGSRTHYADSVKE&PI I BRENSRBFLTOQMSSLRPBDHAVYYCVH 

^^^^ roCS " U ^^ < ^ F ^ Sj<H - -WRO^CitGLBWV SSI S5 ESgTIYYADSVKGRFriEBDMl^r.V^^t s»^.~w-. 0 

t ^ VESGt>3VV Q pGSSL6L; ^^ _WVgQMGgGLSWV AVTST DGSWKTTADSVKtB tFTI ERIJK3KJm J YUJ«HSLHAECTJ<VYTrAfl 

^vEscs^voFGaiz^^ 



o 

^ i 5 !?^ b l raCt L The i >reSen ! 1 inventi ° n relales to ™ vel meth ods for generating variant proteins with increased host string content, and 



WO 2005/056759 A2 IIIllllllllfllHIIIlllllllllI 



SE, SI, SK, TR), OAPI (BF, BJ, CF, CG, CI, CM, GA, GN, For two-letter codes and other abbreviations, refer to the "Guid- 

GQ, GW, ML, MR, NE, SN, TD, TG). ance Notes on Codes and Abbreviations" appearing at the begin- 

Published: nins °f each regular issue of the PCT Gazette. 

— without international search report and to be republished 
upon receipt of that report 



WO 2005/056759 



PCT/US2004/040694 



METHODS OF GENERATING VARIANT PROTEINS WITH INCREASED HOST STRING 
CONTENT AND COMPOSITIONS THEREOF 

[01] This application claims the benefit of under 35 U.S.C. § 1 19(e) to USSNs 
60/527,167, filed December 4, 2003; 60/581,613, filed June 21, 2004; 60/601,665, filed 
August 13, 2004; and, 60/619,483, filed October 14, 2004; all of which are expressly 
incorporated by reference in their entirety. 

FIELD OF THE INVENTION 

[02] The present invention relates to novel methods for generating variant proteins with 
increased host string content, and proteins that are engineered using these methods. 

BACKGROUND OF THE INVENTION 

[03] Many proteins that have the potential to be useful human therapeutics have a 
xenogeneic origin. The use of xenogeneic proteins for therapeutic purposes may be 
advantageous for a variety of reasons, including, for example, the established success of 
hybridoma technology for raising antibodies in rodents, and the possibility of higher efficacy 
with a xenogeneic protein than with a human counterpart. Although xenogeneic proteins are 
a rich source of potential therapeutic molecules, they remain a relatively untapped one. One 
reason for this is that nonhuman proteins are often immunogenic when administered to 
humans, thereby greatly reducing their therapeutic utility. Additionally, even engineered 
proteins of human origin may become immunogenic due to changes in the protein sequence. 

[04] Immunogenicity is the result of a complex series of responses to a substance that is 
perceived as foreign, and may include production of neutralizing and non-neutralizing 
antibodies, formation of immune complexes, complement activation, mast cell activation, 
inflammation, hypersensitivity responses, and anaphylaxis. Several factors can contribute to 
protein immunogenicity, including but not limited to protein sequence, route and frequency of 
administration, and patient population. Immunogenicity may limit the efficacy and safety of a 
protein therapeutic in multiple ways. Efficacy can be reduced directly by the formation of 
neutralizing antibodies. Efficacy may also be reduced indirectly, as binding to either 
neutralizing or non-neutralizing antibodies typically leads to rapid clearance from serum. 
Severe side effects and even death may occur when an immune reaction is raised. One 
special class of side effects results when neutralizing antibodies cross-react with an 
endogenous protein and block its function. 

[05] Because of the clinical success of monoclonal antibodies, immunogenicity reduction 



1 



WO 2005/056759 



PCT/US2004/040694 



of these proteins has been an intense area of investigation. Antibodies are a unique system 
for the development of immunogenicity reduction methods because of the large number of 
highly conserved antibody sequences and the Wealth of high-resolution structural 
information. A number of strategies for reducing antibody immunogenicity have been 
developed. The central aim of all of these approaches has been the reduction of nonhuman, 
and correspondingly immunogenic content, while maintaining affinity for the antigen. 

[06] The dominant method in use for antibody immunogenicity reduction, referred to as 
"humanization", relies principally on the grafting of "donor" (typically mouse or rat) 
complementarity determining regions (CDRs) onto "acceptor" (human) variable light chain 
(VL) and variable heavy chain (VH) frameworks (FRs) (Tsurushita & Vasquez, 2004, 
Humanization of Monoclonal Antibodies, Molecular Biology of B Cells, 533-545, Elsevier 
Science (USA)). This strategy is referred to as "CDR grafting" (Winter US 5225539). 
"Backmutation " of selected acceptor framework residues to the corresponding donor 
residues is often required to regain affinity that is lost in the initial grafted construct (US 
5530101; US 5585089; US 5693761; US 5693762; US 6180370; US 5859205; US 5821337; 
US 6054297; US 6407213). Despite the significant clinical application of antibodies 
engineered using these methods, these methods remain nonrobust with regard to their 
ability to reduce immunogenicity. A number of humanized antibodies have elicited 
substantial immune reaction in clinical studies, with incidences of immune response as high 
as 63 % of patients (Ritter et a/., 2001, Cancer Research 61: 6851-6859). 

[07] The incomplete capacity of current humanization methods for immunogenicity 
reduction are due to significant limitations imposed by the donor-acceptor approach. 
Historically, the use of a single donor has been part of methods aimed at engineering a 
single xenogeneic antibody to be suitable as a human biotherapeutic. However, the use of a 
single acceptor is not required. On the contrary, the use of an acceptor antibody, and the 
use of global homology to select it, place substantial restrictions on the immunogenicity 
reduction process. A principal problem is that the use of overall sequence similarity between 
nonhuman and human sequences as a metric for human immunogenicity is fundamentally 
flawed. This means of measuring the degree of humanness does not accurately account for 
the underlying molecular mechanisms of immune response. The immune system does not 
recognize antigens on the basis of global sequence similarity to human proteins. Rather, 
immune cells, including antigen presenting cells (APCs), T cells, and B cells, recognize 
linear or conformational motifs comprising only a handful of residues. A key step in antigen 
recognition is the formation of peptide-MHC-T cell receptor complexes. APCs express MHC 
molecules that recognize short (approximately nine residue) linear peptide sequences, 



2 



WO 2005/056759 



PCT/US2004/040694 



referred to as MHC agretopes. T cells express T cell receptors that recognize T cell 
epitopes in the context of peptide-MHC complexes. T cells that recognize MHC agretopes 
that are present in human proteins typically undergo apoptosis or become anergic, while T 
cells that recognize foreign agretopes bound to MHC molecules may participate in an 
immune response. Thus the relevant quantity for the immunogenicity of a protein is not its 
global sequence similarity to a human sequence, but rather its sequence content of 
individual human epitopes. 

[08] The donor-acceptor model and the use of global sequence homology that it imposes 
fails in practice. Because CDRs are treated as inviolable, structural incompatibilities are 
introduced at the CDR-FR boundaries. Grafting of foreign donor CDRs onto a human 
acceptor framework creates a substantial number of nonhuman epitopes in each variable 
chain, including not only the epitopes in the foreign CDRs, but also the large number of 
epitopes at the FR-CDR boundaries. This FR-CDR incompatibility is evident when one 
backs away from global homology and looks at more local sequence homologies. CDR 
grafting generally maximizes the donor-acceptor homology of the frameworks at the expense 
of the CDRs (Clark, 2000, Immunology Today 21: 397-402). Ironically this frequently results 
in lower global homology to human antibodies. In reality, the "cut and paste" approach to 
imparting the functional determinants of a nonhuman antibody onto the framework of a 
human one is unnecessary, as careful analysis of the antigen binding determinants of 
antibodies shows that, in fact, the majority of CDR residues are not involved in binding 
antigen (MacCallum et a/., 1996, J. Mol. Biol. 262: 732-745). FR-CDR incompatibility causes 
not only immunological problems at the sequence level, but also causes conformational 
problems at the structural level. As a result, humanization methods based on CDR grafting 
often result in antigen affinity losses of 10-100 -fold, necessitating backmutation to donor 
residues within the framework. This process of backmutation is a hallmark of essentially all 
current humanization efforts, and because it introduces yet additional nonhuman epitopes, 
highlights the inefficiency of these methods;. 

[09] Methods that take an immune epitope approach to reducing antibody immunogenicity 
have been explored (US 5712120; US 2003/0153043). Central to these methods is the 
determination of sequences within a xenogeneic antibody that are in fact immunogenic 
epitopes. Different methods for determination of immunogenicity both theoretical and 
experimental have been described and include determination of potential for amphipathic 
helix formation, binding to MHC, reactivity in a T-cell activation assay. A distinguishing 
feature between these strategies and the present invention is that the present invention 
makes no presumption as to the immunogenicity of specific epitopes. Rather, the primary 



3 



WO 2005/056759 



PCT/US2004/040694 



goal is to maximize the content of human linear sequence strings in the xenogeneic antibody 
as determined by comparison to an alignment of human sequences. The relevant sequence 
dataset comprises strings that are nonimmunogenic for all relevant reasons, including lack of 
interaction with MHC, lack of interaction with T cell receptor, lack of proper processing 
necessary for presentation, and tolerance. 

[10] It is noted that the methods described in US 5712120 and US 2003/0153043 suffer 
additionally in that they fail to address a significant concern for local level sequence 
engineering, namely the requirement for maintaining protein structure, stability, solubility, 
and function. Thus, although the sequence string approach to immunogenicity reduction is 
more accurate than CDR grafting, it will be optimal when coupled with protein design 
methodology that takes into account both local sequence content and conformational 
compatibility at the local and global structural level. In addition to providing scoring functions 
for assessing host string content, the present invention also describes scoring functions that 
evaluate other relevant properties of a protein that may be employed for the simultaneous 
immunogenicity reduction and structural and functional optimization of proteins. 

[111 In summary, the donor-acceptor model imposes significant restrictions on the 
immunogenicity reduction process. With regard to sequence, global sequence homology is 
an inappropriate metric for immunogenicity. With regard to structure, backmutations are 
needed to repair conformational incompatibilities, thereby creating or reintroducing 
nonhuman epitopes. The present invention describes a novel method for antibody 
immunogenicity reduction that steps outside of the donor-acceptor model, and thus the 
sequence and structural restrictions it imposes. The central strategy of the described 
method is that it maximizes the content of human linear sequence strings. In this way 
immunogenicity is addressed at the local sequence level, typically by utilizing the local 
sequence information contained in an alignment of human sequences. This strategy not 
only provides a more accurate measure of the immunogenicity, it enables substitutions to be 
designed in a forward rather than backward manner to repair problems introduced by the 
graft In effect, by addressing immunogenicity at the local sequence string level, the optimal 
balance between binding determinants and humanness can be designed. 

[12] The present invention describes a novel method for reducing the immunogenicity of 
proteins that leverages the nonimmunogenic information contained in natural human 
sequences to score protein sequences for immunogenic content at the sequence string 
level. Furthermore, the described method capitalizes on recent advances in computational 
sequence and structure-based protein engineering methods to quantitatively and 



4 



WO 2005/056759 



PCT/US2004/040694 



systematically determine the optimal balance between human sequence content and protein 
functionality. Because of the wealth of human sequence information available for the 
immunoglobulin protein family, application to human antibodies is emphasized. Applications 
to other proteins are also possible. 

SUMMARY OF THE INVENTION 

[13] The invention disclosed herein provides a novel method for reducing the 
immunogenicity of a protein, wherein the method maximizes the content of sequence strings. 
In a preferred embodiment, the method of the present invention maximizes the content of 
human sequence strings. 

[14] It is an object of the present invention to provide scoring functions that may be used 
to evaluate the human sequence string content of a protein. In a preferred embodiment, the 
scoring function compares the similarity of strings In a protein sequence to the strings that 
compose a set of natural protein sequences. In another preferred embodiment, the set of 
sequences is an aligned set of germline sequences. In additional preferred embodiments, 
the set of sequences contains mature sequences. In the most preferred embodiments, the 
sequences are human sequences. 

[15] It is an object of the present invention to provide scoring functions that may be used 
to evaluate the structural and/or functional fitness of a protein. 

[16] It is an object of the present invention to provide protein variants of a parent protein 
that are engineered using the methods described herein. In a preferred embodiment, the 
parent protein is an immunoglobulin. 

[17] It is an object of the present invention to provide experimental methods for screening 
and testing the protein variants of the present invention. 

[18] The present invention provides isolated nucleic acids encoding the protein variants 
described herein. The present invention provides vectors comprising the nucleic acids, 
optionally, operably linked to control sequences. The present invention provides host cells 
containing the vectors, and methods for producing and optionally recovering the protein 
variants. 

[19] The present invention provides compositions comprising the protein variants 
described herein, and a physiologically or pharmaceutical^ acceptable carrier or diluent. 

[20] The present invention provides novel antibodies and Fc fusions that comprise the 
protein variants disclosed herein. The novel antibodies and Fc fusions may find use in a 



5 



WO 2005/056759 



PCT/US2004/040694 



therapeutic product. 

[21] The present invention provides therapeutic treatment and diagnostic uses for the 
protein variants disclosed herein. 

BRIEF DESCRIPTION OF THE DRAWINGS 

[22] Figure 1 . Human germ line sequences and diversity. The sequences that are known 
to encode the human VH chains (Figure 1a), human VL kappa chains (Figure 1b), and VH 
and VL kappa J chains (Figure 1c) are shown. The VL lambda germline sequences are not 
provided. The germline sequences are numbered according to the numbering scheme of 
Kabat (Kabat et a/., 1991 , Sequences of Proteins of Immunological Interest, 5th Ed., United 
States Public Health Service, National Institutes of Health, Bethesda). The regions of the 
variable region are indicated above the numbering in Figures 1a and 1b, and these include 
framework regions 1 through 3 and the CDRs 1 through 3. Positions that make up the Kabat 
CDRs are underlined. The germline chains are grouped into 7 subfamilies for V H and and 6 
subfamilies for V L , as is known in the art, and these subfamilies are grouped together and 
separated by a blank line. The sequences of the five germlines that make up the IgG light 
kappa J chains (IGKJ1 - IGKJ5), and the six germlines that make up the IgG heavy J chains 
(IGHJ1 - IGHJ5) are shown in Figure 1c. The kappa and lambda light J chains combine 
with the VL/c and Vbl germlines respectively to form the light chain variable region, and the 
heavy J chains combine with the VH germlines and heavy diversity (D) germlines (not 
shown) to form the heavy chain variable region. The V H CDR3 is not part of the V H germ 
line, and is encoded by the D and J genes. 

[23] Figures 2. The quantities described by equations 1, 2, and 3 are illustrated. In 
Figure 2a, IDstring (Equation 1) is illustrated for the string beginning at position i = 15, 
comparing a region of the murine antibody m4D5 VH sequence (VH_m4D5) as parent 
sequence s with the homologous region from the VH human germline sequence (VH_1-2) as 
human sequence h. Only 30 residues from each sequence are shown, and the residues that 
compose the relevant string are bolded. In Figure 2b, IDmax (Equation 2) is illustrated for 
the parent sequence s string that begins at position i = 15 (shown in bold) and the 
homologous regions from an aligned set of 7 VH human germline sequences. In Figure 2c, 
HSC(s) (Equation 3) is illustrated for all strings (i=1 to i=22) in the parent sequence s and the 
homologous regions from an aligned set of 7 VH human germline sequences. 

[24] Figure 3. Sequence, host string content, and structure of WT AC 10 VL. Figure 3a 
shows the sequence of the WT AC10 VL. Figure 3b shows the identity of each residue in 
WT AC 10 VL as compared to the corresponding residue in each sequence of the human 



6 



WO 2005/056759 



PCTAJS2004/040694 



VUc germline. The black horizontal lines delineate the 7 different subfamilies as presented 
in Figure 1, and the black vertical lines delineate the different framework and CDR regions of 
the domain (in the order FR1-CDR1-FR2-CDR2-FR3-CDR3-FR4). A grey square indicates 
that the germline sequence has the same amino acid identity to the residue at the 
corresponding position in the WT AC10 VL sequence. A white square indicates that the two 
sequences differ at that position. Figure 3c shows the continuous 8- and 9-mer strings 
between WT AC 10 VL and each sequence of the human VL/c germline. The black horizontal 
and vertical lines are as described in Figure 3b. A grey square indicates that the germline 
sequence comprises an 9-mer string centered on that position that is an 8 out of 9 or 9 out of 
9 identical match to the corresponding string (centered on the corresponding residue) in the 
WT AC10 VL sequence. Figure 3d shows the structure of the modeled WT AC10 variable 
region. The light chain is shown as grey ribbon, the heavy chain is shown as black ribbon, 
and the CDR residues are indicated as black lines. 

[25] Figure 4. Sequence, host string content, and structure of WT AC10 VH. The figure 
is as described in the figure legend for Figure 3, except that here the light chain is shown as 
black ribbon and the heavy chain is shown as grey ribbon. 

[26] Figure 5. Sequence, host string content, and structure of CDR grafted AC10 VL. 
CDR grafted AC10 VL was derived from the CDRs of WT AC10 and the frameworks of the 
human germline sequence vlk_4-1. Differences between CDR grafted AC10 VL and WT 
AC10 VL are shown as bolded residues in the sequence in Figure 5a, and as black ball and 
sticks in Figure 5d. 

[27] Figure 6. Sequence, host string content, and structure of CDR grafted AC10 VH. 
CDR grafted AC10 VH was derived from the CDRs of WT AC10 and the frameworks of the 
human germline sequence vh_1-3 and substitutions Q108L and A1 13S (Kabat numbering) in 
FR4. 

[28] Figure 7. AC10 VL and VH variants with optimized HSC. AC10 VL and VH variants 
with optimized HSC. The nonredundant set of output sequences from the calculations 
described in Example 1 are shown. For each iteration (Iter) the following are provided: the 
Structural Consensus; Structural Precedence; Human String Content (HSC); Human String 
Similarity (HSS); N 9 max; the Framework Region Homogeneity (FRH); and, the number of 
mutations from WT (Muts). The output sequences were clustered based on their mutational 
distance from the other sequences in the set. These clusters are delineated by the 
horizontal black lines. The "Cluster" column provides this mutational distance quantitatively. 
Differences between the parent WT AC 10 sequence are shown in grey. Positions are 



7 



WO 2005/056759 PCT/US2004/040694 



numbered according to the Kabat numbering scheme, provided at the top. The light grey 
regions bracketed by the black horizontal lines indicate residues in or proximal to the Kabat 
defined CDRs that were masked in the calculation. Sequence differences from WT C225 VL 
are shown in dark grey. 

[29] Figure 8. Sequence, host string content, and structure of L1 AC1 0 VL. 

[30] Figure 9. Sequence, host string content, and structure of L2 AC10 VL. 

[31] Figure 1 0. Sequence, host string content, and structure of L3 AC10 VL. 

[32] Figure 11 . Sequence, host string content, and structure of H1 AC10 VH. 

[33] Figure 1 2. Sequence, host string content, and structure of H2 AC1 0 VH. 

[34] Figure 1 3. Sequence, host string content, and structure of H3 AC1 0 VH. 

[35] Figures 14. AlphaScreen™ assay measuring binding between AC10 variants and 
the target antigen CD30. In the presence of competitor variant antibody, a characteristic 
inhibition curve is observed as a decrease in luminescence signal. The binding data were 
normalized to the maximum and minimum luminescence signal for each particular curve, 
provided by the baselines at low and high antibody concentrations respectively. The curves 
represent the fits of the data to a one site competition model using nonlinear regression, and 
the fits provide IC50s for each antibody. 

[36] Figure 15. Figure 1 1 . SPR sensorgrams showing binding of AC10 WT and variant 
full length antibodies to the CD30 target antigen. The curves consist of an association 
phase and dissociation phase, the separation being marked by a little spike on each curve. 

[37] Figure 16. AlphaScreen™ assay measuring binding between AC10 variants and 
human V1 58 FcKRHIa. 

[38] Figure 17. Cell-based ADCC assay of WT and AC10 variants. Purified human 
peripheral blood monocytes (PBMCs) were used as effector cells, L540 Hodgkin's 
lymphoma cells were used as target cells, and lysis was monitored by measuring LDH 
activity using the Cytotoxicity Detection Kit (LDH, Roche Diagnostic Corporation, 
Indianapolis, IN ). Samples were run in triplicate to provide error estimates (n=3, +/- S.D.). 
Figure 17 shows the dose dependence of ADCC at various antibody concentrations, and the 
curves represent the fits of the data to a sigmoidal dose-response model using nonlinear 
regression. Raw data are presented in Figures 17a and 17b, whereas in Figure 17c the data 
were normalized to a percentage scale of maximal cytotoxicity determined by Triton-X100 
lysis of target cells. 



8 



WO 2005/056759 



PCT/US2004/040694 



[39] Figure 18. Cell-based assay measuring ADCC capacity of WT (H0/L0) and H3/L3 
AC10 antibodies comprising Fc variants that provide enhanced effector function. Raw data 
were normalized to a percentage scale of maximal cytotoxicity determined by Triton-X100 
lysis of target cells. 

[40] Figure 19. AlphaScreen™ assay measuring binding between select H3L3 secondary 
AC10 variants and the target antigen CD30. 

[41] Figure 20. Sequence, host string content, and structure of L3.71 AC10 VL. 

[42] Figure 21 . Sequence, host string content, and structure of L3.72 AC10 VL 

[43] Figure 22. Sequence, host string content, and structure of H3.68AC10VH. 

[44] Figure 23. Sequence, host string content, and structure of H3.69 AC10 VH. 

[45] Figure 24. Sequence, host string content, and structure of H3.70 AC10 VH. 

[46] Figure 25. Amino acid sequences of a AC10 variant antibodies comprising the L3.71 
AC10 variant VL with the CLD constant light chain (Figure 25a) and the H3.70 AC10 variant 
VH with IgG constant chains (Figures 25b - 25e) that may comprise amino acid 
modifications in the Fc region. Figure 25b provides an lgG1 heavy chain with positions that 
may be mutated designated in bold as X 1t X 2 , X 3 , and X4, referring to residues S239, V264, 
A330, and I332. Figure 25c provides one example of a heavy chain described in Figure 25b, 
here comprising the H3.70 AC10 variant VH region with the S239D/A330L/I332E lgG1 
constant region. Figure 25d provides an lgG2 heavy chain with positions that may be 
mutated and designated in bold as X 1t X 2 , X 3 , X4, Z 1f Z 2 , Z3, Z4, and Z 5 referring to residues 
S239, V264, A330, 1332, P233, V234, A235, -236, and G237 (here -236 refers to a deletion 
at EU index position 236). Figure 25e provides one example of a heavy chain described in 
Figure 25d, here comprising the H3.70 AC10 variant VH region with the 
S239D/A330L/I332E/P233E/V234L/A235L/-236G lgG2 constant region. . 

[47] Figure 26. Sequence, host string content, and structure of WT C225 VL. 

[48] Figure 27. Sequence, host string content, and structure of WT C225 VH. 

[49] Figure 28. Sequence, host string content, and structure of CDR grafted C225 VL, 
which was derived from the CDRs of WT C225 and the frameworks of the human germline 
sequence vik_6D-21 and an L106I (Kabat numbering) substitution in FR4. 

[50] Figure 29. Sequence, host string content, and structure of CDR grafted C225 VH, 
which was derived from the CDRs of WT C225 and the frameworks of the human germline 
sequence vh_4-30-4 and an A1 13S (Kabat numbering) substitution in FR4. 



9 



WO 2005/056759 



PCTAJS2004/040694 



[51] Figure 30. C225 VL and VH variants with optimized HSC. The nonredundant set of 
output sequences from the calculations described in Example 2 are shown. 

[52] Figure 31 . C225 VL and VH variants with optimized HSC. The nonredundant set of 
output sequences from the calculations described in Example 2 are shown. 

[53] Figure 32. Sequence, host string content, and structure of 12 C225 VL. 

[54] Figure 33. Sequence, host string content, and structure of L3 C225 VL. 

[55] Figure 34. Sequence, host string content, and structure of L4 C225 VL. 

[56] Figure 35. Sequence, host string content, and structure of H3 C225 VH. 

[57] Figure 36. Sequence, host string content, and structure of H4 C225 VH. 

[58] Figure 37. Sequence, host string content, and structure of H5 C225 VH. 

[59] Figure 38. Sequence, host string content, and structure of H6 C225 VH. 

[60] Figure 39. Sequence, host string content, and structure of H7 C225 VH. 

[61] Figure 40. Sequence, host string content, and structure of H8 C225 VH. 

[62] Figure 41 , SPR sensorgrams showing binding of full length antibody C225 variants 
to the EGFR target antigen. The sensorgrams show binding of C225 WT (L0/H0) and 
variant (L0/H3, L0/H4, L0/H5, L0/H6, L0/H7, L0/H8, L2/H3, L2/H4, L2/H5, L2/H6, L2/H7, 
L2/H8, L3/H3, L3/H4, L3/H5, L3/H6, L3/H7, L3/H8, L4/H3, L4/H4, L4/H5, L4/H6, L4/H7, and 
L4/H8)full length antibodies to the EGFR sensor chip. The curves consist of an association 
phase and dissociation phase, the separation being marked by a little spike on each curve. 

[63] Figures 42. Cell-based ADCC assay of C225 WT (L0/H0) and variant (L0/H3, L0/H4, 
L0/H5, L0/H6, L0/H7, L0/H8, L2/H3, L2/H4, L2/H5, L2/H6, L2/H7, L2/H8, L3/H3, L3/H4, 
L3/H5, L3/H6, L3/H7, L3/H8, L4/H3, L4/H4, L4/H5, L4/H6, L4/H7, and L4/H8) full length 
antibodies. Purified human peripheral blood monocytes (PBMCs) were used as effector 
cells, A431 epidermoid carcinoma cells were used as target cells at a 10:1 effectontarget cell 
ratio, and lysis was monitored by measuring LDH activity using the Cytotoxicity Detection Kit 
(LDH, Roche Diagnostic Corporation, Indianapolis, IN ). Samples were run in triplicate to 
provide error estimates (n=3, +/- S.D.). Figure 42 shows the dose dependence of ADCC at 
various antibody concentrations, normalized to the minimum and maximum levels of lysis for 
the assay. The curves represent the fits of the data to a sigmoidal dose-response model 
using nonlinear regression. 

[64] Figure 43. Sequence, host string content, and structure of WT ICR62 VL. 



10 



WO 2005/056759 



PCTAJS2004/040694 



[65] Figure 44. Sequence, host string content, and structure of WT ICR62 VH. 

[66] Figure 45. Sequence, host string content, and structure of CDR grafted ICR62 VL. 
CDR grafted ICR62 VL was derived from the CDRs of WT ICR62 and the frameworks of the 
human germline sequence vlk_1-17 and an L106I (Kabat numbering) substitution in FR4. 

[67] Figure 46. Sequence, host string content, and structure of CDR grafted ICR62 VH. 
CDR grafted ICR62 VH was derived from the CDRs of WT ICR62 and the frameworks of the 
human germline sequence vhjl-f and substitutions A107T and S108L (Kabat numbering) in 
FR4. 

[68] Figure 47. ICR62 VL and VH variants with optimized HSC. 

[69] Figure 48. Sequence, host string content, and structure of L3 ICR62 VL. 

[70] Figure 49. Sequence, host string content, and structure of H9 ICR62 VH. 

[71] Figure 50. Sequence, host string content, and structure of H10 ICR62 VH. 

[72] Figure 51 . Comparison of VH sequences humanized by the methods in the prior art 
versus the present method. Prior art antibodies include Ctm01 , A5B7, Zenapax, MaE1 1 f 
1129, MHM2, H52, Huzaf, Hu3S193, D3H44, AQC2, 2C4, D3H44, Hfe7A, 5C8, m4D5, 
A.4.6.1, Campath, HuLys11, A.4.6.1, Mylotarg, MEDI-507, huH65_vh, EP-5C7, 9F3, HPC4, 
38C2, Br96, 1A6, and 6.7. Sequences designed using the present invention, including AC10 
H1, H2, and H3 f C225 H3, H4, H5, H6 f H7 ? and H8, and ICR62 H9 and H10, are offset to the 
right. Figure 51a provides the host string content (HSC) as defined by equation 3, Figure 
51b provides the exact string content (ESC) as defined by equation 3a, and Figure 51c 
provides the framework region homogeneity (FRH) as defined by equation 10. Window size 
w was 9 for all calculations. 

[73] Figure 52. Comparison of VL sequences humanized by the methods in the prior art 
versus the present method. Prior art antibodies include Ctm01 , A5B7, Zenapax, MaE1 1 , 
1129, MHM2, H52, Huzaf, Hu3S193, D3H44, AQC2, 2C4, D3H44, Hfe7A, 5C8, m4D5, 
A.4.6.1, Campath, HuLys11, A.4.6.1, Mylotarg, MEDI-507, huH65_vh, EP-5C7, 9F3, HPC4, 
38C2, Br96, 1 A6, and 6.7. Sequences designed using the present invention, including AC10 
L1, L2, and L3, C225 L2, L3, L4, and ICR62 L2, are offset to the right. Figure 52a provides 
the host string content (HSC) as defined by equation 3, Figure 52b provides the exact string 
content (ESC) as defined by equation 3a, and Figure 52c provides the framework region 
homogeneity (FRH) as defined by equation 10. Window size w was 9 for all calculations. 

DETAILED DESCRIPTION OF THE INVENTION 



11 



WO 2005/056759 



PCTAJS2004/040694 



Definitions 

[74] In order that the invention may be more completely understood, several definitions 
are set forth below. Such definitions are meant to encompass grammatical equivalents. 

[75] By " amino acid" as used herein is meant one of the 20 naturally occurring amino 
acids or any non-natural analogues that may be present at a specific, defined position. 

[76] By " amino acid modification " herein is meant an amino acid substitution, insertion, 
and/or deletion in a polypeptide sequence. The preferred amino acid modification herein is a 
substitution. 

[77] By " amino acid substitution " or "substitution" herein is meant the replacement of an 
amino acid at a given position in a protein sequence with another amino acid. 

[78] By " antibody " herein is meant a protein consisting of one or more proteins 
substantially encoded by all or part of the recognized immunoglobulin genes. The 
recognized immunoglobulin genes, for example in humans, include the kappa (/c), lambda 
(/I), and heavy chain genetic loci, which together comprise the myriad variable region genes, 
and the constant region genes mu (p), delta (6), gamma (y) 9 sigma (a), and alpha {a) which 
encode the IgM, IgD, IgG, IgE, and IgA isotypes respectively. Antibody herein is meant to 
include full length antibodies and antibody fragments, and may refer to a natural antibody 
from any organism, an engineered antibody, or an antibody generated recombinantly for 
experimental, therapeutic, or other purposes. By " IgG " as used herein is meant a protein 
belonging to the class of antibodies that are substantially encoded by a recognized 
immunoglobulin gamma gene. In humans this class comprises IgGt, lgG2, lgG3, and lgG4. 

[79] By " corresponding " or " eguivalenf residues as meant herein are residues that 
represent similar or homologous sequence and/or structural environments between a first 
and second protein, or between a first protein and set of multiple proteins. In order to 
establish homology, the amino acid sequence of a first protein is directly compared to the 
sequence of a second protein. After aligning the sequences, using one or more of the 
homology alignment programs known in the art (for example using conserved residues as 
between species), allowing for necessary insertions and deletions in order to maintain 
alignment (i.e., avoiding the elimination of conserved residues through arbitrary deletion and 
insertion), the residues equivalent to particular amino acids in the primary sequence of the 
first protein are defined. Alignment of conserved residues preferably should conserve 100% 
of such residues. However, alignment of greater than 75% or as little as 50% of conserved 
residues is also adequate to define equivalent residues. Corresponding residues may also 



12 



WO 2005/056759 



PCT/US2004/040694 



be defined by determining structural homology between a first and second protein that is at 
the level of tertiary structure for proteins whose structures have been determined. In this 
case, equivalent residues are defined as those for which the atomic coordinates of two or 
more of the main chain atoms of a particular amino acid residue of the proteins (N on N, CA 
on CA, C on C and O on O) are within 0.13 nm and preferably 0.1 nm of each other after 
alignment. Alignment is achieved after the best model has been oriented and positioned to 
give the maximum overlap of atomic coordinates of non-hydrogen protein atoms of the 
proteins. 

[80] By "CDR " as used herein is meant a Complementarity Determining Region of an 
antibody variable domain. Systematic identification of residues included in the CDRs have 
been developed by Kabat (Kabat et ah, 1991 , Sequences of Proteins of Immunological 
Interest, 5th Ed., United States Public Health Service, National Institutes of Health, 
Bethesda) and alternately by Chothia (Chothia & Lesk, 1987, J. Mol. Biol. 196: 901-917; 
Chothia et al., 1989, Nature 342: 877-883; Al-Lazikani et al., 1997, J. MoL BioL 273: 927- 
948). For the purposes of the present invention, CDRs are defined as a slightly smaller set 
of residues than the CDRs defined by Chothia. VL CDRs are herein defined to include 
residues at positions 27-32 (CDR1), 50-56 (CDR2), and 91-97 (CDR3), wherein the 
numbering is according to Chothia. Because the VL CDRs as defined by Chothia and Kabat 
are identical, the numbering of these VL CDR positions is also according to Kabat. VH 
CDRs are herein defined to include residues at positions 27-33 (CDR1), 52-56 (CDR2), and 
95-102 (CDR3), wherein the numbering is according to Chothia. These VH CDR positions 
correspond to Kabat positions 27-35 (CDR1 ), 52-56 (CDR2), and 95-1 02 (CDR3). 

[81] By " framework" as used herein is meant the region of an antibody variable domain 
exclusive of those regions defined as CDR's. Each antibody variable domain framework can 
be further subdivided into the contiguous regions separated by the CDR's (FR1, FR2, FR3 
and FR4). 

[82] By "germline" as used herein is meant the set of sequences that compose the natural 
genetic repertoire of a protein, and its associated alleles. 

[83] , By " host" as used herein is meant a family, genus, species or subspecies, group of 
individuals or even a single individual. A host group of individuals can be selected for based 
upon a variety of criteria, such as MHC allele composition, etc. In a preferred embodiment, 
a host is canine, murine, primate, or human. In the most preferred embodiment, a host is 
human. 

[84] By "host string " or " host sequence 0 as used herein is meant a string or sequence that 



13 



WO 2005/056759 



PCT/US2004/040694 



encodes any part of a naturally occurring host protein. 

[85] By " humanized " antibody as used herein is meant an antibody comprising a human 
framework region and one or more CDR's from a non-human (usually mouse or rat) 
antibody. The non-human antibody providing the CDR's is called the " donor and the human 
immunoglobulin providing the framework is called the " acceptor" . One says that the donor 
antibody has been humanized", by the process of "humanization". 

[86] By " identity" as used herein is meant the number of residues in a first sequence that 
are identical to the residues in a second sequence after alignment of the sequences to 
achieve the maximum identity. 

[87] By " immune epitope " or " epitope " herein is meant a linear sequence of amino acids 
that is located in a protein of interest. Epitopes may be analyzed for their potential for 
immunogenicity. Epitopes may be any length, preferably 9-mers. 

[88] By "immunogenicity" herein is meant the ability of a protein to elicit an immune 
response, including but not limited to production of neutralizing and non-neutralizing 
antibodies, formation of immune complexes, complement activation, mast cell activation, 
inflammation, and anaphylaxis. 

[89] By " immunoglobulin do) " herein is meant a protein consisting of one or more proteins 
substantially encoded by immunoglobulin genes. Immunoglobulins include but are not 
limited to antibodies. Immunoglobulins may have a number of structural forms, including but 
not limited to full length antibodies, antibody fragments, and individual immunoglobulin 
domains. By " immunoglobulin (lg) domain " herein is meant a region of an immunoglobulin 
that exists as a distinct structural entity as ascertained by one skilled in the art of protein 
structure. Ig domains typically have a characteristic p-sandwich folding topology. The 
known Ig domains in the IgG class of antibodies are V H , CH1 (Cy1), CH2 (Cy2), CH3 (Cy3), 
V Ll and C L . 

[90] By "natural seouence " or " natural protein" as used herein is meant a protein that has 
been determined to exist absent any experimental modifications. Also included are 
sequences that can be predicted to exist in nature based on experimentally determined 
sequences. An example of such a predicted sequence is an antibody that can be predicted 
to exist based on the established patterns of germline recombination! In this case the large 
size of the predicted antibody repertoire makes the actual experimental determination of all 
mature recombined antibodies not practical. 

[91] By " parent " or " parent protein " as used herein is meant a protein that is subsequently 



14 



WO 2005/056759 



PCT/US2004/040694 



modified to generate a variant. The parent protein may be a naturally occurring protein, or a 
variant or engineered version of a naturally occurring protein. Parent protein may refer to 
the protein itself, compositions that comprise the parent protein, or the amino acid sequence 
that encodes it Accordingly, by " parent antibody" as used herein is meant an antibody that 
is subsequently modified to generate a variant antibody. Accordingly, by " parent sequence" 
as used herein is meant the sequence that encodes the parent protein or parent antibody. 

[92] By " position " as used herein is meant a location in the sequence of a protein. 
Positions may be numbered sequentially, or according to an established format, for example 
Kabat, Chothia, and/or the EU index as in Kabat. 

[93] By "protein" herein is meant at least two covalently attached amino acids, which 
includes proteins, polypeptides, oligopeptides and peptides. The protein may be made up of 
naturally occurring amino acids and peptide bonds, or synthetic peptidomimetic structures. 

[94] By " reduced immunogenicitv" herein is meant a decreased ability to activate the 
immune system, when compared to the parent protein. For example, a protein variant can 
be said to have "reduced immunogenicity" if it elicits neutralizing or non-neutralizing 
antibodies in lower titer or in fewer patients than the parent protein. A protein variant also 
can be said to have "reduced immunogenicity" if it shows decreased binding to one or more 
MHC alleles or if it induces T cell activation in a decreased fraction of patients relative to the 
parent protein. 

[95] By " residue " as used herein is meant a position in a protein and its associated amino 
acid identity. For example, proline 9 (also referred to as Pro9, also referred to as P9) is a 
residue in the WT AC1 0 VH region. 

[96] By " scoring function" herein is meant any equation or method for evaluating the 
fitness of one or more amino acid modifications in a protein. The scoring function may 
involve a physical or chemical energy term, or may involve knowledge-, statistical-, 
sequence-based energy terms, and the like. 

[97] By " string" as used herein is meant a contiguous sequence that encodes any part of 
a protein. Strings may comprise any 2 or more linear residues, with the number of 
contiguous residues being defined by the " window " or " window size ". Window sizes of 2 - 20 
are preferred, with 7-13 more preferred, with 9 most preferred. 

[98] By " target" as used herein is meant the molecule that is bound specifically by a 
protein. A target may be a protein, carbohydrate, lipid, or other chemical compound. The 
target of an antibody is its antigen, also referred to as its target antigen. 



15 



WO 2005/056759 



PCT/US2004/040694 



[99] By "variable region" as used herein is meant the region of an immunoglobulin that 
comprises one or more Ig domains substantially encoded by any of the VL (including V/c and 
VA) and/or V H genes that make up the light chain (including kappa and lambda) and heavy 
chain immunoglobulin genetic loci respectively. A light or heavy chain variable region (VL 
and VH) consists of a "framework" or TFT region interrupted by three hypervariable regions 
referred to as "complementarity determining regions" or "CDRs". The extent of the 
framework region and CDRs have been precisely defined, for example as in Kabat (see 
"Sequences of Proteins of Immunological Interest," E. Kabat et al., U.S. Department of 
Health and Human Services, (1983)), and as in Chothia. The framework regions of an 
antibody, that is the combined framework regions of the constituent light and heavy chains, 
serves to position and align the CDRs, which are primarily responsible for binding to an 
antigen. 

[100] By "variant protein " or " protein variant " . or "variant " as used herein is meant a protein 
that differs from a parent protein by virtue of at least one amino acid modification. Protein 
variant may refer to the protein itself, a composition comprising the protein, or the amino 
sequence that encodes it Preferably, the protein variant has at least one amino acid 
modification compared to the parent protein, e.g. from about one to about ten amino acid 
modifications, and preferably from about one to about five amino acid modifications 
compared to the parent. The protein variant sequence herein will preferably possess at least 
about 80% homology with a parent protein sequence, and most preferably at least about 
90% homology, more preferably at least about 95% homology. Accordingly, by 
" immunoglobulin variant " as used herein is meant an immunoglobulin that differs from a 
parent immunoglobulin by virtue of at least one amino acid modification. 

[101] By "wild type orWT" herein is meant an amino acid sequence or a nucleotide 
sequence that is found in nature and includes allelic variations. A WT protein has an amino 
acid sequence or a nucleotide sequence that has not been intentionally modified. 

[102] The protein variants of the present invention may be derived from parent proteins 
that are themselves from a wide range of sources. The parent protein may be substantially 
encoded by one or more genes from any organism, including but not limited to humans, 
mice, rats, rabbits, camels, llamas, dromedaries, monkeys, preferably mammals and most 
preferably humans and mice and rats. Although in a preferred embodiment the parent 
protein is nonhuman, in some embodiments of the present invention the parent protein may 
be human or similar to human. The parent protein may comprise more than one protein 
chain, and thus may be a monomer or an oligomer, including a homo- or hetero-oligomer. In 



16 



WO 2005/056759 



PCT7US2004/040694 



a preferred embodiment, the parent protein is an antibody, referred to as the parent 
antibody. The parent antibody need not be naturally occurring. For example, the parent 
antibody may be an engineered antibody, including but not limited to nonhuman and 
chimeric antibodies. The parent antibody may be fully human, obtained for example using 
transgenic mice (Bruggemann et a/., 1997, Curr Opin Biotechnol 8:455-458) or human 
antibody libraries coupled with selection methods (Griffiths et a/., 1998, Curr Opin Biotechnol 
9:102-108). The parent antibody need not be naturally occurring. For example, the parent 
antibody may be an engineered antibody, including but not limited to chimeric antibodies and 
humanized antibodies (Clark, 2000, Immunol Today 21:397-402). The parent antibody may 
be an engineered variant of an antibody that is substantially encoded by one or more natural 
antibody genes. In one embodiment, the parent antibody has been affinity matured, as is 
known in the art, or engineered in some other way. The parent antibodies of the present 
invention may be substantially encoded by immunoglobulin genes belonging to any of the 
antibody classes, and may comprise sequences belonging to the IgG class of antibodies, 
including lgG1 , lgG2, lgG3, or lgG4, or alternatively the IgA (including subclasses lgA1 and 
lgA2), IgD, IgE, IgG, or IgM classes of antibodies. 

[103] Virtually any binding partner or antigen may be targeted by the proteins of the 
present invention. A number biotherapeutic proteins and antibodies that are approved for 
use, in clinical trials, or in development may thus benefit from immunogenicity reduction 
methods of the present invention. In a preferred embodiment, the less immunogenic protein 
of the present invention is an antibody. The less immunogenic antibody may comprise 
sequences belonging to the IgG (including lgG1 , lgG2, lgG3, or lgG4), IgA (including 
subclasses lgA1 and lgA2), IgD, IgE, IgG, or IgM classes of antibodies, with the IgG class 
being preferred. The less immunogenic antibodies of the present invention may be full 
length antibodies, or antibody fragments. Constant regions need not be present, but if they 
are, they will likely be substantially identical to human immunoglobulin constant regions. 

[104] The constant region of the antibody may be modified in some way to make it more 
effective therapeutically. For example, the constant region may comprise substitutions that 
enhance therapeutic properties. Most preferred substitutions and optimized effector function 
properties are described in USSN 10/672,280, PCT US03/30249, and USSN 10/822,231, 
and USSN 60/627,774, filed 1 1/12/2004 and entitled "Optimized Fc Variants". Other known 
Fc variants that may find use in the present invention include but are not limited to those 
described in US.6,737,056; PCT US2004/000643; USSN 10/370,749; PCT/US2004/005112; 
US 2004/0132101; USSN 10/672,280; PCT/US03/30249; US 6,737,056, US 2004/0002587; 
WO 2004/063351; Idusogie et al.,.2001, J. Immunology 166:2571-2572; Hinton et al., 2004, 



17 



WO 2005/056759 



PCTAJS2004/040694 



J. Biol. Chem. 279(8): 6213-6216. In alternate embodiments, the constant region may 
comprise one or more engineered glycoforms, as is known in the art (Umana et a/., 1999, 
Nat Biotechnol 17:176-180; Davies ef a/., 2001, Biotechnol Bioeng 74:288-294; Shields et 
a/., 2002, J Biol Chem 277:26733-26740; Shinkawa et a/ M 2003, J Biol Chem 278:3466- 
3473); (US 6,602,684; USSN 10/277,370; USSN 10/113,929; PCT WO 00/61739A1; PCT 
WO 01/29246A1 ; PCT WO 02/31 140A1 ; PCT WO 02/30954A1); (Potelligent™ technology 
[Biowa, Inc., Princeton, NJ]; GlycoMAb™ glycosylation engineering technology [GLYCART 
biotechnology AG, Zurich, Switzerland]). 

[105] The protein variants of the present invention may find use in a wide range of protein 
products. In one embodiment the protein is a therapeutic, a diagnostic, or a research 
reagent, preferably a therapeutic. Alternatively, the protein of the present invention may be 
used for agricultural or industrial uses. In a preferred embodiment, the protein is a 
therapeutic that is used to treat a disease. By "disease " herein is meant a disorder that may 
be ameliorated by the administration of a pharmaceutical composition comprising a protein 
of the present invention. Diseases include but are not limited to autoimmune diseases, 
immunological diseases, infectious diseases, inflammatory diseases, neurological diseases, 
and oncological and neoplastic diseases including cancer. In one embodiment, a protein of 
the present invention is the only therapeutically active agent administered to a patient. 
Alternatively, the protein of the present invention is administered in combination with one or 
more other therapeutic agents, including but not limited to cytotoxic agents, 
chemotherapeutic agents, cytokines, growth inhibitory agents, anti-hormonal agents, kinase 
inhibitors, anti-angiogenic agents, cardioprotectants, or other therapeutic agents. The 
proteins of the present invention may be combined with other therapeutic regimens. For 
example, in one embodiment, the patient to be treated with the protein may also receive 
radiation therapy and/or undergo surgery. In an alternate embodiment, the protein of the 
present invention is conjugated or operably linked to another therapeutic compound. The 
therapeutic compound may be a cytotoxic agent, a chemotherapeutic agent, a toxin, a 
radioisotope, a cytokine, or other therapeutically active agent. In yet another embodiment, a 
protein of the present invention may be conjugated to a protein or molecule for utilization in 
tumor pretargeting or prodrug therapy. Other modifications of the proteins of the present 
invention are contemplated herein. For example, the protein may be linked to one of a 
variety of nonproteinaceous polymers, for example e.g., polyethylene glycol (PEG). 

[106] Pharmaceutical compositions are contemplated wherein a protein of the present 
invention and one or more therapeutically active agents are formulated. Formulations of the 
proteins of the present invention are prepared for storage by mixing the protein having the 



18 



WO 2005/056759 



PCT/US2004/040694 



desired degree of purity with optional pharmaceutical^ acceptable carriers, excipients or 
stabilizers (Remingtons Pharmaceutical Sciences 16th edition, Osol, A. Ed.,1980), in the 
form of lyophilized formulations or aqueous solutions. The formulations to be used for in 
vivo administration are preferably sterile. The proteins disclosed herein may also be 
formulated as immunolipospmes, or entrapped in microcapsules. The concentration of the 
protein of the present invention in the formulation may vary from about 0.1 to 1 00 weight %. 
In a preferred embodiment, the concentration of the protein is in the range of 0.003 to 1.0 
molar. In order to treat a patient, a therapeutically effective dose of the protein of the 
present invention may be administered. The exact dose will depend on the purpose of the 
treatment, and will be ascertainable by one skilled in the art using known techniques. 
Dosages may range from 0.01 to 100 mg/kg of body weight or greater, for example 0.1, 1, 
10, or 50 mg/kg of body weight, with 1 to 10mg/kg being preferred. Administration of the 
pharmaceutical composition comprising a protein of the present invention, preferably in the 
form of a sterile aqueous solution, may be done in a variety of ways, including, but not 
limited to, orally, subcutaneously, intravenously, intranasally, intraotically, transdermal^, 
topically, intraperitoneally, intramuscularly, intrapulmonary, inhalably, vaginally, parenterally, 
rectally, or intraocularly. As is known in the art, the pharmaceutical composition may be 
formulated accordingly depending upon the manner of introduction. 

Description of the Methodology 

[107] The present invention provides a novel method for reducing the immunogenicity of a 
protein. A central principle of the described method is that substitutions are designed to 
maximize the content of human linear sequence strings using an alignment of human 
sequences. For application to antibodies, this approach to immunogenicity reduction 
excludes the use of the single donor-acceptor model employed in humanization methods. 
By stepping outside of the limitations imposed by the need to choose a human acceptor 
sequence a priori, a more immunologically relevant approach to immunogenicity reduction is 
enabled. Sequence information and structural information may be used to score potential 
amino acid substitutions. The scoring results are used to design protein variant libraries, 
which are subsequently screened experimentally to determine favorable substitutions. 
Feedback from experimental data may guide subsequent iterations of design and 
experimental screening, ultimately enabling protein variants to be engineered with the 
optimal balance between biophysical and immunological constraints. 

Sequences 

[108] Central to the method described herein is that a set of host sequences provides 



19 



WO 2005/056759 



PCT/US2004/040694 



information as to the degree to which linear sequence strings have the potential to be 
immunogenic. Thus the set of sequences employed is an important parameter. In the most 
common embodiment, the sequences are a set of human sequences that are homologous in 
sequence and/or structure to the parent sequence. As is known in the art, some proteins 
share a common structural scaffold and are homologous in sequence. This information may 
be used to gain insight into particular positions in the protein family. Sequence alignments 
are often carried out to determine which protein residues are conserved and which are not 
conserved. That is to say, by comparing and contrasting alignments of protein sequences, 
the degree of variability at a position may be observed, and the types of amino acids that 
occur naturally at positions may be observed. Thus for the present invention, typically the 
sequences are aligned such that the conserved or similar residues that exist between the 
parent sequence and the set of human sequences and among the set of human sequences 
can be identified. Methods for sequence alignment are well known in the art, and include 
alignments based on sequence and structural homology. 

[109] Protein sequence information can be obtained, compiled, and/or generated from 
sequence alignments of naturally occurring proteins from any organism, including but not 
limited to mammals. Because a preferred embodiment of present invention is directed 
towards immunogenicity reduction for biotherapeutics, the sequences that compose the set 
are most preferably human. The source of the sequences may vary widely, may be a 
database that is compiled publicly or privately, and may be may include one or more of the 
known general protein and nucleic acid sequences databases, including but not limited to 
SwissProt, GenBank and Entrez, and EMBL Nucleotide Sequence Database. Because a 
preferred embodiment of the present invention is its application to the immunogenicity 
reduction of immunoglobulins, a number of immunoglobulin databases may be useful for 
obtaining sequences, including but not limited to the Kabat database (Johnson & Wu, 2001, 
Nucleic Acids Res 29:205-206; Johnson & Wu, 2000, Nucleic Acids Res 28:214-218), the 
IMGT database (IMGT, the international ImMunoGeneTics information system®; Lefranc et 
a/., 1999, Nucleic Acids Res 27:209-212; Ruiz et a/., 2000 Nucleic Acids Re. 28:219-221; 
Lefranc et a/., 2001 , Nucleic Acids Res 29:207-209; Lefranc et a/., 2003, Nucleic Acids Res 
31 :307-31 0), and VBASE. 

[110] As is well known in the art, immunoglobulins possess a high degree of sequence and 
structural homology, and therefore alignment of sequences provides a wealth of information. 
Due to the existence of deletions and insertions in these alignments, numbering conventions 
have been adopted to enable a normalized reference to conserved positions in 
immunoglobulin families or subfamilies. Those skilled in the art will appreciate that these 



20 



WO 2005/056759 



PCT/US2004/040694 



conventions consist of nonsequential numbering in specific regions of an immunoglobulin 
sequence, and thus accordingly the positions of any given immunoglobulin as defined by any 
given numbering scheme will not necessarily correspond to its sequential sequence or to 
those in an alternate numbering scheme. For all variable regions discussed in the present 
invention, numbering is according to the numbering scheme of Kabat (Kabat et a/., 1991 , 
Sequences of Proteins of Immunological Interest, 5th Ed., United States Public Health 
Service, National Institutes of Health, Bethesda). For all constant region positions discussed 
in the present invention, number is according to the EU index as in Kabat Alternate 
numbering schemes may find use in the present invention, including but not limited that of 
Chothia (Chothia & Lesk, 1987, J.Mol. Biol 196: 901-917; Chothia et al t 1989, Nature 342: 
877-883; Al-Lazikani et a/., 1997, J. Mol Biol 273: 927-948). 

[111] In a most preferred embodiment, the set of human sequences used is an aligned set 
of human germline immunoglobulin sequences. For example, Figures 1a - 1c provide the 
set of sequences that compose the human antibody variable region germline (VH, VL, and J 
chains), along with the corresponding diversity at each position. The human germline 
repertoire for immunoglobulin heavy chain variable regions and immunoglobulin light chain 
kappa variable regions have been reported (Matsuda et al., 1998, J Exp Med 188: 2151- 
2162; Zachau, 2000, Biol Chem 381:951-954; Pallares et al., 1999, Exp Clin Immunogenet 
16(1): 36-60; Barbie & Lefranc, 1998, Exp Clin Immunogenet 15(3): 171-83). The human 
immunoglobulin kappa variable (IGKV) genes and joining (IGKJ) segments. Barbie V, 
Lefranc MP). The rationale for use of this type of sequence information as a metric for 
humanness is that the strings that compose the human germline should be minimally 
immunogenic. Sequences need not be human genomic or germline sequences. In other 
preferred embodiments, human antibody variable region sequences are derived not from 
germline information, but rather from matured antibodies obtained for example from 
hybridoma technology or cDNA libraries. 

[112] For many of the genes in the human immunoglobulin germline, several different 
alleles have been identified. Although the polymorphisms detected in many of the alleles do 
not change the amino acid sequence of the gene, in a great number of cases the sequence 
is changed. In choosing a set of sequences to use in the method described herein, different 
sets of sequences may be chosen. When choosing a single allele as representative of a 
specific gene the most cautious approach is to choose that sequence which is closest to the 
consensus of the entire germline. This subset of sequences would thereby be most likely to 
be represented within the population as a whole. Alternatively, a much greater sequence 
diversity could be sampled by choosing representative sequences that are furthest from the 



21 



WO 2005/056759 



PCT/US2004/040694 



consensus. Another approach yielding greater diversity would be to use multiple alleles 
where they exist for each germiine. At this time, there is little or no quantitative data on 
allele frequency within the population. When allele frequency becomes available, a more 
informed decision can be made regarding the likelihood of tolerance for a specific non- 
consensus allele within the target patient population. 

[113] When two or more possible substitutions are being evaluated for use at a specific 
position when both are found in the human germiine, the decision may become subjective. 
In such a case additional information can be incorporated that may reflect different levels of 
expression of particular genes (Cox et al. Eur J Immunol. 1994 Apr;24(4):827-36). One 
underlying assumption of such a strategy would be that relative expression level of a 
particular germiine (or corresponding sequence strings) correlates with the relative 
immunogenicity. 

[1 14] The sequences used for the method disclosed herein are those of homologous 
proteins with sufficient homology to allow their alignment with the protein whose 
immunogenicity is being reduced. One might argue that if a particular protein sequence is 
found anywhere within the expressed human genome that there is innate tolerance to that 
peptide. Such a proposition greatly increases the number of possible sequences that could 
be used to reduce the immunogenicity of a protein. In such a case however, alignment of 
proteins that are not structurally homologous would likely be prohibitive. In addition, the 
processing of a protein to produce the strings to which tolerance is developed may be 
structurally determined. Therefore, a specific strings may be nonimmunogenic in its native 
context but immunogenic in an altered structural context. 

Scoring Functions - String Content 

[115] In order to evaluate the fitness of protein variants, amino acid modifications in the 
parent protein may be scored using a variety of scoring functions. Central to preferred 
embodiment of immunogenicity reduction method described herein is that at least one 
scoring function is aimed at maximizing the content of host linear sequence strings that are 
present in a set of host sequences. Typically, but not always, a computer is used to score 
potential amino acid substitutions. 

[116] In one embodiment, substitutions may be scored according to their occupancy in the 
set of host sequences, i.e., whether or not a given amino acid is part of the diversity at a 
given position. The use of position-specific alignment information to generate a list of 
considered amino acids at a variable position is well known in the art; see for example 
Lehmann & Wyss, 2001 , Curr Opin Biotechnol 12(4): 371-5; Lehmann et a/., 2000, Biochim 



22 



WO 2005/056759 



PCT/US2004/040694 



BiophysActa 1543(2):408-415; Rath & Davidson, 2000, Protein Sc/, 9(1 2):2457-69; 
Lehmann ef a/., 2000, Protein Eng 13(1):49-57; Desjarlais & Berg, 1993, Proc Natl Acad Sci 
USA 90(6):2256-60; Desjarlais & Berg, 1992, Proteins 12(2):101-4; Henikoff & Henikoff, 
2000, Adv Protein Chem 54:73-97; Henikoff & Henikoff, 1 994, J Mol Biol 243(4);574-8. 
Thus, for example, for the parent nonhuman VUc sequence aligned to the human sequences 
in Figure 1b, substitutions to be considered at position 1 would be Ala, Asp, Glu, Asn, and 
Val. In a more preferred embodiment, substitutions are scored based on their frequency in 
the set of human sequences listed. For example, in the previous example, Asp and Glu 
occur most frequently at position 1, and thus may be more preferable substitutions that Ala, 
Asn, or Val. The basis for this scoring function is that the frequency of a given amino acid at 
a given position in the alignment is proportional to its potential for being in a host string. 

[117] Occupancy and frequency provide relatively straightforward approximations for 
designing substitutions that have the potential for reduced immunogenicity. Their use, 
however, does not take into account the context of the parent sequence. Although 
frequency is proportional to the potential for a substitution to increase host content of a 
string, it is not a direct measure. In order to more accurately incorporate the information 
present in an aligned set of host sequences into a measure of immunogenicity, an approach 
can be taken wherein the linearity or contiguity of a given position in the context of the 
strings that comprise it is considered. In this most preferred embodiment, substitutions in a 
parent sequence are scored based on the probability of removing a nonhost string and 
replacing it with a less immunogenic string, namely one present in the set of host 
sequences. This method of scoring may employ the calculation of identity or percent identity 
of a parent string to a host string within a window of equivalent positions. In one 
embodiment, the identity of a string in sequence s to a host string in sequence h, (IDstring), 
can be presented as the sum of amino acid sequence identities in a given window size, 
according to equation 1: 

Equation 1 IDstring (i 9 w) = atjh 

where w is the string window size, i is the first position in the string, aa s j is the amino acid at 
position j of sequence s, aa h j is the amino acid at position j of the host sequence h, and the 
Kronecker delta function is used to return a value of 1 for a match (for example if the parent 
and host amino acids at position j are both serine) and 0 if there is no match (for example if . 
the parent amino acid at position j is a serine but the host amino acid is a leucine). Figure 
2a illustrates equation 1 using a region of the VH of murine anti-Her2 antibody m4D5 



23 



WO 2005/056759 



PCT/US2004/040694 



(VH_m4D5) as the parent sequence s and the homologous region from the VH human 
germline (VHM-2) as human sequence h. 

[118] In a further embodiment, it is assumed that the most immunologically appropriate 
measure of host string content at position i is the maximal identity between a string of 
sequence s and any host sequence in the alignment, as calculated in equation 2: 

Equation 2 /Z)max = max(Y 8 . >) 

where HS is the set of host sequences. In other words, if IDstring at position i is equal to w 
for any one of the host sequences, IDmax = w as well, and the i th string is assumed to be 
minimally immunogenic. The concept of the IDmax quantity represented by Equation 2 is 
illustrated in Figure 2b. 

[119] Finally, these equations can be combined to calculate a single numerical metric for 
total host string content (HSC) of a sequence s by summing the IDmax values over all 
pertinent sequence positions, as in equation 3: 

Equation 3 HSC{s) = 100 Y max( Y 8 , h ) 

where L is the length of the sequence and HS is the set of host sequences in the alignment. 
A perfectly host sequence would have an HSC of 100. One might alternatively say that such 
a sequence is 100% host. The concept of the HSC quantity represented by Equation 3 is 
illustrated in Figure 2c. In alternative embodiments, Equation 3 can be modified further such 
that the final score is dependent on the relative usage of each host sequence in the 
alignment. Strings from sequences that are more frequently expressed by hosts are 
expected to be more tolerized, and therefore may be given correspondingly higher influence 
in a scoring system. 

[120] In an alternative embodiment, one can measure the exact string content (ESC) as in 
Equation 3a: 

Equation 3a ESC(s) = 100 : Y max 8 . * , 

(Z-w+1) j~{ h z H s ^^^^i 

where the notation aa 5 LI+w _i refers the contiguous sequence string in protein s from position i 
to position i+w-1. In this embodiment, only perfect matches of size w are counted in the 
score. 

[121] It is worth noting that, since the scoring systems in Equations 3 and 3a are based on 



24 



WO 2005/056759 



PCT/US2004/040694 



local sequence identity and/or similarity evaluated over windows of defined size, a sequence 
with high HSC can be constructed of sequence segments that are maximally similar to 
different members of the set of host sequences at different positions. 

[122] The above measure of hostness is likely to be more immunologically relevant than 
the more commonly used global identity measure of equation 4: 



1 1 

Equation 4 globallD = 100 -max— Y 8 , b 



Equation 4 disregards the extent of contiguous sequence identity, which is particularly 
relevant for capturing the molecular behavior of the immune system. 

[123] Additional scoring functions similar to equations 3 are also possible. For example, as 
will be appreciated by those skilled in the art, there is some uncertainty regarding the 
hostness of a string wherein IDmax = w-1, w-2, etc. In one alternative embodiment, 
sequence similarity is compared instead of identity, using any of a variety of amino acid 
substitution matrices (e.g. PAM, BLOSUM62, etc.), providing a host string similarity (HSS) 
score as in equation 5: 



Equations HSS(s) = 100 Y maxfe** J J ) 

L-W+l j% heHS J 

where S is a substitution score comparing any two amino acids. In yet another alternative, 
sequence identities are weighted according to the extent of identity, as in equation 6: 

Equation 6 HSC(s) = 1 00 ■ — - Y /(max( Y S , , )) 

(L-w+l) tt °°>' aa > 

where f is a continuous or noncontiguous function dependent on IDmax. For example, 
perfect matches can be weighted greater than near perfect matches {e.g. f(w)=1 , f(w-1 )=.5, 
etc.), and poor matches can be discarded (e.g. f(w-3)=f(w-2) = 0). 

String window size 

[124J The fundamental binding units of class I and class II MHC proteins are both 9 amino 
acids. In a preferred embodiment, the window size w used to create and score parent 
sequences is 9. However, it is also known that additional peptide flanking residues (PFRs) 
can influence T-cell recognition (via the TCR) of class II MHC-peptide complexes (see for 
example Arnold et al., 2002, J Immunology 169(2): 739-49), with the residues at positions P- 
1 (one position before the 1 st MHC binding position) and P1 1 being most influential. 



25 



WO 2005/056759 



PCT/US2004/040694 



Because these effects might influence immune tolerance, a desirable goal of the invention, 
larger window sizes (e.g. 1 2) can be used. It should be noted however, that sequences 
optimized with similar window sizes are highly correlated. 

Optimization of HSC 

[125] Although a definition of string scoring systems is useful, an efficient process for 
discovering sequences with high HSC is also desirable. It is therefore a further aspect of the 
invention to provide methods for dynamic optimization of HSC given the described scoring 
systems. 

[1 26] Desirable features of an optimization method include but are not limited to the 
following: 1) the output sequences are optimal or near-optimal (subject to design constraints) 
in their host string content; 2) structural constraints can be used to modulate the nature of 
the optimized sequences; and 3) multiple near-optimal solutions can be generated. 
Additionally, in some preferred embodiments, host string content may be maximized using a 
minimal number of substitutions. 

[1 27] In a preferred embodiment, an iterative algorithm for optimization of HSC works as 
follows. 1) a parent sequence and set of host sequences are defined; 2) mutational 
constraints are defined at functionally or structurally important positions, referred to herein as 
masking - in a preferred embodiment, for antibody applications, positions within or 
structurally proximal to CDR residue (as defined by herein, or alternatively as defined by 
Kabat or Chothia) and/or interface are masked, locked, or fixed so that mutations are not 
possible (in some embodiments this constraint can be relaxed if the potential mutation is a 
conservative substitution of the parent amino acid). In a preferred embodiment, positions 
within 5 angstroms of a CDR residue or interface are masked. In other preferred 
embodiments, positions within 6.5 angstroms of a CDR residue or interface are masked; 3) 
host sequence segments (up to a defined length: lengths from 1-6 are typical) are collected 
from the alignment and stored for each position: segments that violate the mutational 
constraints are not collected; 4) each segment is analyzed for its potential impact on HSC, in 
the context of the current parent sequence, defined as String Impact (SI) in equation 7: 

Equation 7 & ~+ < z » = HSC ^ )) ~ HSC{parenf) ^ 

where y m (z) is a host segment of length z replacing segment x at position m, and s(y m ) and 
. parent are versions of the parent sequence that include these segments (the parent 
sequence contains xjz)). 5) a single string is randomly selected from all stored host strings; 
the probability of selection is biased and proportional to the impact on HSC and inversely 



26 



WO 2005/056759 



PCT/US2004/040694 



proportional to the number of mutations relative to the current parent sequence, as in 
Equation 8: 

Paz sr(*M-+yM) 

Equation 8 **** 

This selected string is substituted into the current parent sequence for its corresponding 
parent amino acids on an amino acid string by amino acid string basis. This kind of selection 
bias tends to optimize host string content with minimal perturbation of the original sequence. 
6) steps 4 and 5 are repeated until no further optimization is possible (no segment 
substitutions have a favorable impact on host string content). 

[128] Such an algorithm is inherently non-deterministic, so independent runs of the 
algorithm will tend to generate different solutions (this is a favorable feature). In a preferred 
embodiment, such an algorithm is applied numerous times to generate a diverse array of 
unique solutions. These solutions can be further clustered such that representative 
sequences can be prioritized for further analysis. For example, in one embodiment, the 
solution sequences are clustered into groups of similar sequences according to mutational 
distance, using a nearest neighbor single linkage hierarchical clustering algorithm to assign 
sequences to related groups based on similarity scores. Clustering algorithms may be 
useful for classifying sequences into representative groups. Representative groups may be 
defined, for example, by similarity. Measures of similarity include, but are not limited to 
sequence similarity and energetic similarity. Thus the output sequences from computational 
screening may be clustered around local minima, referred to herein as clustered sets of 
sequences. Sets of sequences that are close in sequence space may be distinguished from 
other sets. In one embodiment, diversity across clustered sets of sequences may be 
sampled by experimentally testing only a subset of sequences within each clustered set. For. 
example, all or most of the clustered sets could be broadly sampled by including the lowest 
energy sequence from each clustered set of sequences to be experimentally tested. 
Because the sequence space of solutions with optimized HSC can be large, additional 
methods can be applied to ensure that a broad set of sequences is created. In a preferred 
embodiment, individual framework sequences generated by the procedure are clustered 
separately to generate a list of nonredundant basis framework regions (FRs) with high HSC. 
These basis FRs are then computationally assembled in all combinations along with the 
CDRs to generate a secondary list of solution sequences (which will usually have some 
overlap with the primary set). Alternatively, the basis FRs may be combined into an 



27 



WO 2005/056759 



PCTAJS2004/040694 



experimental library, for example a combinatorial library. 
Framework Diversity 

[129] Application of this algorithm will generate variant protein solutions for which HSC is 
higher than the original parent sequence. It will also frequently generated solutions in which 
substituted strings are derived from different members of the alignment. The variant 
sequences derived using the present invention generally have unique properties relative to 
sequences generated using other methodologies. For example, in the context of an 
antibody, the protein variants of the invention frequently derive their host string content from 
a combination of different host germline sequences. This may be true even within a single 
FR. Quantification of these properties is useful for defining the nature of sequences derived 
using the present invention. A clear distinction emerges from a comparison of exact string 
content (meaning a perfect match over window w) in any single germline sequence versus 
exact w-mer string content within the set of all germline sequences (content of strings for 
which IDmax = w). Single germline exact string content (SGESC) of a variant sequence v 
may thus be defined as: 

Equation 9 SGESCCv) = 100 max Y 8 

M 



[130] This quantity provides the extent to which a string-optimized sequence has string 
identity with the closest single germline sequence. Using this definition, it is also possible to 
assess the extent to which the high host string content of a given variant sequence v is 
derived from a single germline as opposed to multiple germline sequences. Framework 
region homogeneity (FRH) is defined as follows: 



Equation 1 0 FRH(v) = - , ^ , 

w ESC(y) 1 ^ 



x max Y 5 „ h 
_ SGESC(y) _ heHs ^ °°L^,™L^x 

Smax£ , „ 



[131] In other words, if a variant sequence's exact string content is derived solely from a 
single germline sequence, the FRH would be close to 1.0. It should be noted that a similar 
or identical quantity can be defined for non-antibody proteins. Alternatively, as is the case 
with many of the variant sequences created by the present invention, FRH values can be 
significantly less than 1, with values ranging from 0.4 to 1.0, indicating, as expected, that 
sequences with high exact string content can be discovered with contributions from multiple 



28 



WO 2005/056759 



PCTAJS2004/040694 



germline subfamilies and sequences. As described more fully in Example 5 below, variant 
sequences generated using the present invention have high HSC values yet many have low 
FRH values, indicating their HSC is derived from multiple germline frameworks. 

Additional Scoring 

[132] The above methods of scoring use the information present in an aligned set of host 
sequences as a metric of irnmunogenicity to maximize the content of host linear sequence 
strings in a parent sequence. In addition to such scoring functions, other scoring functions 
and methods may be employed. Such additional scoring functions may be aimed at the 
same goal as the aforementioned linear string scoring function, namely irnmunogenicity 
reduction of the parent protein. Alternatively, such additional scoring functions may be used 
to achieve other goals, for example optimization of protein stability, solubility, expression, 
pharmacokinetics, and/or aspects of protein function such as affinity of the parent protein for 
a target ligand, specificity, effector function, and/or enzymatic activity. For example an 
additional scoring function may be employed to enhance the affinity of an antibody variable 
domain for its target antigen. Such additional scoring functions may be employed statically 
or dynamically for the generation of optimized protein variants. A number of embodiments 
are described below as preferred additional scoring functions that may be used with the 
aforementioned linear string scoring method of the present invention. However, these are 
not meant to constrain the invention to these embodiments, and it should be clear that any 
method of scoring the fitness of an amino acid modification in a parent protein may be 
coupled with the novel linear string scoring method of the present invention so that optimal 
protein variants may be designed. 

[133] In a preferred embodiment, substitutions are scored based on their structural 
compatibility with the structure of the parent protein. Such methods of scoring may require 
the structural coordinates that describe the three-dimensional structure of the protein, for 
example as obtained by X-ray crystallographic and nuclear magnetic resonance (NMR) 
techniques. Suitable proteins structures may also be obtained from structural models, which 
may be generated by methods that are known in the art of structural biology, including but 
not limited to de novo and homology modeling. Structure-based scoring functions may 
include any number of potentials that describe or approximate physical or chemical energy 
terms, including but not limited to a van der Waals potential, a hydrogen bond potential, an 
atomic solvation potential or other solvation models, a secondary structure propensity 
potential, an electrostatic potential, a torsional potential, an entropy potential, and/or 
additional energy terms. In other preferred embodiments, scoring methods may also be 



29 



WO 2005/056759 PCT/US2004/040694 



derived from sequence information, including but not limited to knowledge-based potentials 
derived from protein sequence and/or structure statistics, threading potentials, reference 
energies, pseudo energies, homology-based energies, and sequence biases derived from 
sequence alignments. In alternately preferred embodiments, both structural and sequence- 
based potentials are used to generate one or more scoring functions that may be coupled 
with the linear string scoring method of the present invention. 

[134] In a most preferred embodiment, a scoring method is used wherein the structural and 
functional integrity of substitutions are evaluated using a sequence and structure-based 
scoring function described in USSN 60/528,229, filed December 8, 2003, entitled Protein 
Engineering with Analogous Contact Environments; and USSN 60/602,566, filed August 17, 
2004, entitled Protein Engineering with Analogous Contact Environments. This method 
combines sequence alignment information and structural information to predict the structural 
compatibility of one or more substitutions with a protein structure template. Nearest 
neighbor structure-based scores generated by this method include Structural Consensus 
and Structural Precedence as provided in the Examples. This method is particularly well 
suited for application to evaluating the structural fitness of immunoglobulins due to their 
substantial sequence and structural homology. 

[135] In a preferred embodiment, substitutions are scored using a scoring function or 
computational design program that is substantially similar to Protein Design Automation® 
(PDA®) technology, as is described in US 6,188,965; US 6,269,312; US 6,403,312; US 
6,708,120; US 6,804,61 1; US 6,792,356; USSN 09/782,004; USSN 09/812,034; USSN 
09/927,790; USSN 10/218,102; USSN 10/101,499; USSN 10/218,102; USSN 10/666,311; 
USSN 10/665,307; USSN 10/888,748; PCT WO 98/07254; PCT WO 99/24229; PCT WO 
01/40091; and PCT WO 02/25588. In another preferred embodiment, a computational 
design method substantially similar to Sequence Prediction Algorithm™ (SPA™) technology 
is used, as is described in (Raha et a/., 2000, Protein ScL 9: 1106-1119), USSN 09/877,695, 
and USSN 10/071,859. In another preferred embodiment, the computational methods 
described in USSN 10/339,788, are used. 

[136] In another preferred embodiment, optimized sequences are also assessed for 
surface similarity with host antibodies. Ensuring similarity may be important for reducing the 
probability of introducing novel 3D epitopes, which are potentially recognized by B-cell 
receptors. In a preferred embodiment, surface similarity at position i is quantified as follows: 

Equation 11 surfscoreii) = maxe Jm} 

keHS 



30 



WO 2005/056759 



PCT/US2004/040694 



where fj exp is the fraction accessibility of position i to solvent, proximity(i j) is the spatial 
proximity of positions i and j in the three-dimensional structure of the protein, S is a measure 
of amino acid similarity, and T is a temperature factor used to tune the stringency of the 
similarity comparison. It will be appreciated from the equation that if sequences are identical 
in the region of position i, a surfscore of 1.0 will be approached. Alternatively, a score of 1.0 
can also be achieved if a position is completely buried (i.e. fi exp =0), since the position would 
not be accessible to B-cell receptors. Lower scores represent surface positions for which 
there are significant differences between the variant sequence and the most similar host 
sequence. In a preferred embodiment, the proximity between two positions is inversely 
related to their distance (e.g. a Gaussian or exponential function of the distance), and the 
proximity of a position to itself is 1 .0. In a preferred embodiment, the decay of the proximity 
function is tuned such that patches of positions correspond to the size of a typical antibody 
epitope. , 

[137] Other surface properties may also be desirable. For example, optimized variant 
sequences may be assessed for the exposure of nonpolar amino acids, which is generally 
expected to decrease solubility. In such cases, variant sequences with lower nonpolar 
exposure can be prioritized over alternatives. Surface electrostatic properties may also be 
assessed for variant sequences. In a preferred embodiment, surface properties are 
assessed for multiple variants with optimized HSC, in order to limit the set of variants that 
will be experimentally screened. 

[138] In another embodiment, substitution matrices or other knowledge-based scoring 
methods are used to identify alternate sequences that are likely to retain the structure and 
function of the protein. Such scoring methods can be used to quantify how conservative a 
given substitution or set of substitutions is. In most cases, conservative mutations do not 
significantly disrupt the structure and function of proteins (see for example, Bowie et a/., 
1990, Science 247: 1306-1310, Bowie & Sauer, 1989, Proc. Nat Acad, ScL USA 86: 2152- 
2156, and Reidhaar-Olson & Sauer, 1990, Proteins 7: 306-316). However, non-conservative 
mutations can destabilize protein structure and reduce activity (see for example, Lim et a/., 
1992, Biochem. 31: 4324-4333). Substitution matrices including but riot limited to 
BLOSUM62 provide a quantitative measure of the compatibility between a sequence and a 
target structure, which can be used to predict non-disruptive substitution mutations (Topham 
et a/., 1997, Prot Eng. 10: 7-21). The use of substitution matrices to design peptides with 
improved properties has been disclosed (Adenot etal., 1999, J. Mol. Graph. Model. 17: 292- 
309). Substitution matrices include, but are not limited to, the BLOSUM matrices (Henikoff & 
Henikoff, 1992, Proc. Nat. Acad. Sci. USA 89: 10917, the PAM matrices, the Dayhoff matrix, 



31 



WO 2005/056759 



PCT/US2004/040694 



and the like. For a review of substitution matrices, see for example Henikoff, 1 996, Curr. 
Opin. Struct Biol. 6: 353-360. It is also possible to construct a substitution matrix based on 
an alignment of a given protein of interest and its homologs; see for example Henikoff & 
Henikoff, 1996, Comput. Appl. BioscL 12: 135-143. 

[139] In a preferred embodiment, other methods for scoring immunogenicity may 
additionally be used. Most preferably, immunogenicity may be scored using a function that 
considers peptide binding to one or more MHC molecules. For example, substitutions would 
be scored such that there are no or a minimal number of immune epitopes that are predicted 
to bind, with high affinity, to any prevalent MHC alleles. These methods of scoring may be 
useful, for example, for designing substitutions in VH CDR3, for which scoring using human 
germline strings may be less straightforward. Several methods of identifying MHC-binding 
epitopes in protein sequences are known in the art and may be used to score epitopes in an 
antibody. See for example WO 98/52976; WO 02/079232; WO 00/3317; USSN 09/903,378; 
USSN 10/039,170; USSN 60/222,697; USSN 10/339788; PCTWO 01/21823; and PCT WO 
02/00165; Mallios, 1999, Bioinformatics 15: 432-439; Mallios, 2001, Bioinformatics 17: 942- 
948; Stumiolo et a/., 1999, Nature Biotech. 17: 555-561; WO 98/59244; WO 02/069232; WO 
02/77187; Marshall etal., 1995, J. Immunol 154: 5927-5933; and Hammer et aA, 1994, J. 
Exp. Med. 180: 2353-2358. Sequence-based information can be used to determine a 
binding score for a given peptide - MHC interaction (see for example Mallios, 1999, 
Bioinformatics 15: 432-439; Mallios, 2001 , Bioinformatics 17: p942-948; Sturniolo et. a/., 
1999, Nature Biotech. 17: 555-561). It is possible to use structure-based methods in which a 
given peptide is computationally placed in the peptide-binding groove of a given MHC 
molecule and the interaction energy is determined (for example, see WO 98/59244 and WO 
02/069232). Such methods may be referred to as "threading" methods. Alternatively, purely 
experimental methods can be used; for example a set of overlapping peptides derived from 
the protein of interest can be experimentally tested for the ability to induce T-cell activation 
and/or other aspects of an immune response, (see for example WO 02/77187). In a 
preferred embodiment, MHC-binding propensity scores are calculated for each 9-residue 
frame along the protein sequence using a matrix method (see Sturniolo et at, supra] 
Marshall et a/., 1995, J. Immunol. 154: 5927-5933, and Hammer et a/., 1994, J. Exp. Med. 
180: 2353-2358). It is also possible to consider scores for only a subset of these residues, 
or to consider also the identities of the peptide residues before and after the 9-residue frame 
of interest. The matrix comprises binding scores for specific amino acids interacting with the 
peptide binding pockets in different human class II MHC molecule. In the most preferred 
embodiment, the scores in the matrix are obtained from experimental peptide binding 



32 



WO 2005/056759 



PCT/US2004/040694 



studies. In an alternate preferred embodiment, scores for a given amino acid binding to a 
given pocket are extrapolated from experimentally characterized alleles to additional alleles 
with identical or similar residues lining that pocket. Matrices that are produced by 
extrapolation are referred to as "virtual matrices". 

[140] In alternate embodiments, additional scoring functions are employed that predict 
reactive sites within a protein, such as deamidation sites, glycosylation sites, oxidation sites, 
protealytic cleavage sites, and the like. 

[141] It will be appreciated by one of skill in the art that the use of combinations of any of 
the aforementioned scoring functions and/or other scoring functions is contemplated. In one 
embodiment, this could be accomplished by evaluating the outputs of the results from 
separate calculations. Alternatively, scoring functions may be combined into one scoring 
term. This latter strategy enables different scoring terms to be weighted separately, thus 
providing more control over the relative contributions of the scoring terms, and a greater 
capacity to tune the scoring function for a desired engineering strategy. 

Additional Optimization of Sequences 

[142] In a preferred embodiment, after optimized protein variants have been engineered 
using the aforementioned scoring functions, additional optimization of protein variants may 
be carried out. In this way, an optimized protein variant can be thought of as primary variant 
or template for further optimization, and variants of this primary variant can be thought of as 
secondary variants. Because variant sequences of the invention are preferably derived from 
a HSC-increasing procedure in which substitution of structurally important positions is 
disallowed or discouraged (for example masking), it is likely that additional optimization of 
HSC is possible if those positions are allowed to vary in a secondary analysis. Optimization 
of other properties is possible, including but are not limited to protein affinity, expression, 
specificity, solubility, activity, and effector function. Thus the variant sequences derived in 
the primary analysis can represent variants for further optimization. In a preferred 
embodiment, the secondary analysis comprises the steps of 1) string analysis of the 
template sequence to identify secondary amino acid diversity that will have neutral, positive, 
or minimal impact on HSC; 2) experimental production of secondary variant sequences 
using the diversity derived in step 1 ; and 3) experimental screening of secondary variant 
sequences. 

[143] For these purposes, the string impact (SI) of a single substitution at position m from 
amino acid x to amino acid y can be quantified as in Equation 7 (where segment length z = 
1). As will be appreciated, the maximum possible string increase (for a single substitution at 



33 



WO 2005/056759 



PCTAJS2004/040694 



an internal position) is w and the maximum possible decrease is w. 

[144] In a most preferred embodiment, substitutions in a primary variant are chosen as 
those substitutions that will result in zero or positive string impact, referred to herein as string 
neutral and string positive substitutions respectively. In other embodiments, substitutions 
that result in negative string impact, i.e. string negative substitutions, may also be 
considered for engineering secondary variants. Secondary variants then may be 
constructed, expressed, and tested experimentally. Secondary substitutions that show 
favorable properties with respect to antigen affinity, effector function, stability, solubility, 
expression, and the like, may be combined in subsequent variants to generate a more 
optimized therapeutic candidate. 

Optimization of Non-Xenogeneic Proteins 

[145] It can be appreciated that the optimization described above is not restricted to 
immunogenicity reduction of xenogeneic proteins. Evaluation of potential substitutions for 
string impact provides an excellent strategy for generating substitution diversity for 
engineering protein variants with optimized properties. A clear advantage of this approach is 
that it generates protein variants with minimal immunogenicity risk, the importance of which 
has been discussed extensively and is a primary goal of the present invention. An additional 
advantage of this approach is that because the sequences being used to evaluate string 
impact are typically derived from a set of naturally evolved host sequences, variants 
designed are effectively enriched for stability, solubility, and other favorable properties. The 
utility of this capability lies in the fact that there are innumerable amino acid modifications 
that are detrimental or deleterious to proteins. By screening a quality set of variant diversity, 
the chances are increased that a protein variant of the desired property will be obtained. 
The capacity of the string impact approach to generate a quality set of variant diversity 
derives from the greater tolerance to mutation of positions which sample greater diversity, 
and the greater propensity of amino acids in a set of naturally evolved sequences to be 
compatible with a homologous protein's structure, stability, solubility, function, and the like. 

[146] This string impact approach to variant design may be applied not only to the 
generation of secondary variants as described above, but may also be used to engineer 
amino acid modifications in proteins that are presumably already minimally immunogenic. 
This may include, for example, natural host proteins. Alternatively, and in a preferred 
embodiment, the string impact strategy may be applied to engineer modifications in an 
antibody variable region (VH or VL) that is humanized (Clark, 2000, Immunol Today 21 :397- 
402); or "fully human" as obtained for example using transgenic mice (Bruggemann et a/., 



34 



WO 2005/056759 



PCT/US2004/040694 



1997, Curr Opin Biotechnol 8:455-458) or human antibody libraries coupled with selection 
methods (Griffiths et a/., 1998, Curr Opin Biotechnol 9:102-108). As with optimization of 
primary variant sequences described above, the string impact analysis described here can 
be used to identify secondary diversity that will have neutral, positive, or minimal impact on 
HSC, as well as potentially other favorable properties. Such diversity can then be used to 
screen for optimized versions of these sequences without increasing the risk of 
immunogenicity. 

Experimental Production, Screening, and Testing 

[147] Methods for production and screening of protein variants are well known in the art. 
General methods for antibody molecular biology, expression, purification, and screening are 
described in Antibody Engineering, edited by Duebel & Kontermann, Springer-Verlag, 
Heidelberg, 2001 ; and Hayhurst & Georgiou, 2001 , Curr Opin Chem Biol 5:683-689; 
Maynard & Georgiou, 2000, Annu Rev Biomed Eng 2:339-76. Also see the methods 
described in USSN 10/339788, filed on March 3, 20O3, USSN 10/672,280, filed September 
29, 2003, and USSN 1 0/822,231 , filed March 26, 2004. 

[148] |n one embodiment of the present invention, the library sequences are used to create 
nucleic acids that encode the member sequences, and that may then be cloned into host 
cells, expressed and assayed, if desired. These practices are carried out using well-known 
procedures, and a variety of methods that may find use in the present invention are 
described in Molecular Cloning - A Laboratory Manual, 3 rd Ed. (Maniatis, Cold Spring Harbor 
Laboratory Press, New York, 2001), and Current Protocols in Molecular Biology (John Wiley 
& Sons). The nucleic acids that encode the protein variants of the present invention may be 
incorporated into an expression vector in order to express the protein. Expression vectors 
typically comprise a protein operably linked, that is placed in a functional relationship, with 
control or regulatory sequences, selectable markers, any fusion partners, and/or additional 
elements. The protein variants of the present invention may be produced by culturing a host 
cell transformed with nucleic acid, preferably an expression vector, containing nucleic acid 
encoding the protein variants, under the appropriate conditions to induce or cause 
expression of the protein. A wide variety of appropriate host cells may be used, including 
but not limited to mammalian cells, bacteria, insect cells, and yeast. For example, a variety 
of cell lines that may find use in the present invention are described in the ATCC cell line 
catalog, available from the American Type Culture Collection. The methods of introducing 
exogenous nucleic acid into host cells are well known in the art, and will vary with the host 
cell used. 



35 



WO 2005/056759 



PCT/US2004/040694 



[149] In a preferred embodiment, protein variants are purified or isolated after expression. 
Proteins may be isolated or purified in a variety of ways known to those skilled in the art. 
Standard purification methods include chromatographic techniques, electrophoretic, 
immunological, precipitation, dialysis, filtration, concentration, and chromatofocusing 
techniques. As is well known in the art, a variety of natural proteins bind antibodies, for 
example bacterial proteins A, G, and L, and these proteins may find use in the present 
invention for purification. Purification can often be enabled by a particular fusion partner. 
For example, proteins may be purified using glutathione resin if a GST fusion is employed, 
Nf 2 affinity chromatography if a His-tag is employed, or immobilized anti-flag antibody if a 
flag-tag is used. For general guidance in suitable purification techniques, see Protein 
Purification: Principles and Practice, 3 rd Ed., Scopes, Springer-Verlag, NY, 1994. 

[150] Protein variants may be screened using a variety of methods, including but not 
limited to those that use in vitro assays, in vivo and cell-based assays, and selection 
technologies. Automation and high-throughput screening technologies may be utilized in the 
screening procedures. Screening may employ the use of a fusion partner or label, for 
example an immune label, isotopic label, or small molecule label such as a fluorescent or 
colorimetric dye. 

[151] In a preferred embodiment, the functional and/or biophysical properties of protein 
variants are screened in an in vitro assay. In a preferred embodiment, the protein is 
screened for functionality, for example its ability to catalyze a reaction or its binding affinity to 
its target. Binding assays can be carried out using a variety of methods known in the art, 
including but not limited to FRET (Fluorescence Resonance Energy Transfer) and BRET 
(Bioluminescence Resonance Energy Transfer) -based assays, AlphaScreen™ (Amplified 
Luminescent Proximity Homogeneous Assay), Scintillation Proximity Assay, ELISA 
(Enzyme-Linked Immunosorbent Assay), SPR (Surface Plasmoh Resonance, also known as 
BIACORE®), isothermal titration calorimetry, differential scanning calorimetry, gel 
electrophoresis, and chromatography including gel filtration. These and other methods may 
take advantage of some fusion partner or label. Assays may employ a variety of detection 
methods including but not limited to chromogenic, fluorescent, luminescent, or isotopic 
labels. The biophysical properties of proteins, for example stability and solubility, may be 
screened using a variety of methods known in the art. Protein stability may be determined 
by measuring the thermodynamic equilibrium between folded and unfolded states. For 
example, protein variants of the present invention may be unfolded using chemical 
denaturant, heat, or pH, and this transition may be monitored using methods including but 
not limited to circular dichroism spectroscopy, fluorescence spectroscopy, absorbance 



36 



WO 2005/056759 



PCT/US2004/040694 



spectroscopy, NMR spectroscopy, calorimetry, and proteolysis. As will be appreciated by 
those skilled in the art, the kinetic parameters of the folding and unfolding transitions may 
also be monitored using these and other techniques. The solubility and overall structural 
integrity of a protein variant may be quantitatively or qualitatively determined using a wide 
range of methods that are known in the art. Methods which may find use in the present 
invention for characterizing the biophysical properties of protein variants include gel 
electrophoresis, chromatography such as size exclusion chromatography and reversed- 
phase high performance liquid chromatography, mass spectrometry, ultraviolet absorbance 
spectroscopy, fluorescence spectroscopy, circular dichroism spectroscopy, isothermal 
titration calorimetry, differential scanning calorimetry, analytical ultra-centrifugation, dynamic 
light scattering, proteolysis, and cross-linking, turbidity measurement, filter retardation 
assays, immunological assays, fluorescent dye binding assays, protein-staining assays, 
microscopy, and detection of aggregates via ELISA or other binding assay. Structural 
analysis employing X-ray crystallographic techniques and NMR spectroscopy may also find 
use. 

1152) In a preferred embodiment, protein variants are screened using one or more cell- 
based or in vivo assays. For such assays, purified or unpurified proteins are typically added 
exogenously such that cells are exposed to individual variants or pools of variants belonging 
to a library. These assays are typically, but not always, based on the function of the protein; 
that is, the ability of the protein to bind to its target and mediate some biochemical event, for 
example effector function, ligand/receptor binding inhibition, apoptosis, and the like. Such 
assays often involve monitoring the response of cells to the protein, for example cell survival, 
cell death, change in cellular morphology, or transcriptional activation such as cellular 
expression of a natural gene or reporter gene. For example, such assays may measure the 
ability of antibody variants to elicit ADCC, ADCP, or CDC. For some assays additional cells 
or components, that is in addition to the target cells, may need to be added , for example 
serum complement, or effector cells such as peripheral blood monocytes (PBMCs), NK cells, 
macrophages, and the like. Such additional cells may be from any organism, preferably 
humans, mice, rat, rabbit, and monkey. Proteins may cause apoptosis of certain cell lines 
expressing the target, or they may mediate attack on target cells by immune cells which 
have been added to the assay. Methods for monitoring cell death or viability are known in 
the art, and include the use of dyes, immunochemical, cytochemical, and radioactive 
reagents. For example, caspase staining assays may enable apoptosis to be measured, 
and uptake or release of radioactive substrates or fluorescent dyes such as alamar blue may 
enable cell growth or activation to be monitored. In a preferred embodiment, the DELFIA® 



37 



WO 2005/056759 



PCTAJS2004/040694 



EuTDA-based cytotoxicity assay (Perkin Elmer, MA) is used. Alternatively, dead or 
damaged target cells may be monitored by measuring the release of one or more natural 
intracellular proteins, for example lactate dehydrogenase. Transcriptional activation may 
also serve as a method for assaying function in cell-based assays. In this case, response 
may be monitored by assaying for natural genes or proteins which may be upregulated, for 
example the release of certain interleukins may be measured, or alternatively readout may 
be via a reporter construct. Cell-based assays may also involve the measure of 
morphological changes of cells as a response to the presence of a protein. Cell types for 
such assays may be prokaryotic or eukaryotic, and a variety of cell lines that are known in 
the art may be employed. Alternatively, cell-based screens are performed using cells that 
have been transformed or transfected with nucleic acids encoding the variant proteins. That 
is, protein variants are not added exogenously to the cells. For example, in one 
embodiment, the cell-based screen utilizes cell surface display. A fusion partner can be 
employed that enables display of variants on the surface of cells (Witrrup, 2001 , Curr Opin 
> Biotechnol, 12:395-399). 

[153] As is known in the art, a subset of screening methods are those that select for 
favorable members of a library. The methods are herein referred to as " selection methods ", 
and these methods find use in the present invention for screening protein variants. When 
protein libraries are screened using a selection method, only those members of a library that 
are favorable, that is which meet some selection criteria, are propagated, isolated, and/or 
observed. As will be appreciated, because only the most fit variants are observed, such 
methods enable the screening of libraries that are larger than those screenable by methods 
that assay the fitness of library members individually. Selection is enabled by any method, 
technique, or fusion partner that links, covalently or noncovalently, the phenotype of a 
protein with its genotype, i.e., the function of a protein with the nucleic acid that encodes it 
For example the use of phage display as a selection method is enabled by the fusion of 
library members to the gene III protein. In this way, selection or isolation of protein variants 
that meet some criteria, for example binding affinity to the protein's target, also selects for or 
isolates the nucleic acid that encodes it. Once isolated, the gene or genes encoding variants 
may then be amplified. This process of isolation and amplification, referred to as panning, 
may be repeated, allowing favorable protein variants in the library to be enriched. Nucleic 
acid sequencing of the attached nucleic acid ultimately allows for gene identification. 

[154] A variety of selection methods are known in the art that may find use in the present 
invention for screening protein libraries. These include but are not limited to phage display 
(Phage display of peptides and proteins: a laboratory manual, Kay et ai t 1996, Academic 



38 



WO 2005/056759 



PCT/US2004/040694 



Press, San Diego, CA, 1996; Lowman etal., 1991, Biochemistry 30:10832-10838; Smith, 
1985, Science 228:1315-1317) and its derivatives such as selective phage infection 
(Malmborg et al. y 1997, J Mol Biol 273:544-551), selectively infective phage (Krebber et a/., 

1997, J Mol Biol 268:619-630), and delayed infectivity panning (Benhar et a/., 2000, J Mol 
Biol 301 :893-904), cell surface display (Witrrup, 2001, Curr Opin Biotechnol, 12:395-399) 
such as display on bacteria (Georgiou et al., 1997, Nat Biotechnol 15:29-34; Georgiou et al., 

1993, Trends Biotechnol 1 1 :6-10; Lee et al., 2000, Nat Biotechnol 18:645-648; Jun ef al., 

1998, Nat Biotechnol 16:576-80), yeast (Boder & Wittrup, 2000, Methods Enzymol 328:430- 
44; Boder & Wittrup, 1997, Nat Biotechnol 1 5:553-557), and mammalian cells (Whitehorn ef 
al., 1995, BioAechnology 13:1215-1219), as well as in vitro display technologies (Amstutz ef 
al., 2001 , Curr Opin Biotechnol 12:400-405) such as polysome display (Mattheakis et al., 

1994, Proc Natl Acad Sci USA 91:9022-9026), ribosome display (Hanes et al., 1997, Proc 
Natl Acad Sci USA 94:4937-4942), mRNA display (Roberts & Szostak, 1997, Proc Natl Acad 
Sci USA 94:12297-12302; Nemoto etal., 1997, FEBS Lett 414:405-408), and ribosome- 
inactivation display system (Zhou etal., 2002, J Am Chem Soc 124, 538-543). 

[1 55] Other selection methods that may find use in the present invention include methods 
that do not rely on display, such as in vivo methods including but not limited to periplasmic 
expression and cytometric screening (Chen ef ai, 2001 , Nat Biotechnol 19:537-542), the 
protein fragment complementation assay (Johnsson & Varshavsky, 1994, Proc Natl Acad Sci 
USA 91 :10340-10344; Pelletier ef al., 1998, Proc Natl Acad Sci USA 95:12141-12146), and 
the yeast two hybrid screen (Fields & Song, 1989, Nature 340:245-246) used in selection 
mode (Visintin etal., 1999, Proc Natl Acad Sci USA 96:11723-11728). In an alternate 
embodiment, selection is enabled by a fusion partner that binds to a specific sequence on 
the expression vector, thus linking covalently or noncovalently the fusion partner and 
associated variant library member with the nucleic acid that encodes them. For example, 
USSN 09/642,574; USSN 10/080.376; USSN 09/792,630; USSN 10/023,208; USSN 
09/792,626; USSN 10/082,671; USSN 09/953,351; USSN 10/097,100; USSN 60/366,658; 
PCT WO 00/22906; PCT WO 01/49058; PCT WO 02/04852; PCT WO 02/04853; PCT WO 
02/08023; PCT WO 01/28702; and PCT WO 02/07466 describe such a fusion partner and 
technique that may find use in the present invention. In an alternative embodiment, in vivo 
selection can occur if expression of the protein imparts some growth, reproduction, or 
survival advantage to the cell. 

[1 56] A subset of selection methods referred to as "directed evolution" methods are those 
that include the mating or breading of favorable sequences during selection, sometimes with 
the incorporation of new mutations. As will be appreciated by those skilled in the art, 



39 



WO 2005/056759 



PCT/US2004/040694 



directed evolution methods can facilitate identification of the most favorable sequences in a 
library, and can increase the diversity of sequences that are screened. A variety of directed 
evolution methods are known in the art that may find use in the present invention for 
screening protein variants, including but not limited to DNA shuffling (PCT WO 00/42561 A3; 
PCT WO 01/70947 A3), exon shuffling (US 6,365,377; Kolkman & Stemmer, 2001, Nat 
Biotechnol 19:423-428), family shuffling (Crameri et a/., 1998, Nature 391:288-291; US 
6,376,246), RACHITT™ (Coco et a/., 2001 , Nat Biotechno\ 19:354- 359; PCT WO 02/06469), 
STEP and random priming of in vitro recombination (Zhao ef a/., 1998, Nat Biotechnol 
16:258-261; Shao et a/., 1998, Nucleic Acids Res 26:681-683), exonuclease mediated gene 
assembly (US 6,352,842; US 6,361,974), Gene Site Saturation Mutagenesis™ (US 
6,358,709), Gene Reassembly™ (US 6,358,709), SCRATCHY (Lutz et a!., 2001, Proc Natl 
Acad Sci USA 98:1 1248-1 1253), DNA fragmentation methods (Kikuchi et a/., Gene 236:159- 
167), single-stranded DNA shuffling (Kikuchi et a/., 2000, Gene 243:133-137), and 
AMEsystem™ directed evolution protein engineering technology (Applied Molecular 
Evolution) (US 5,824,514; US 5,817,483; US 5,814,476; US 5,763,192; US 5,723,323). 

[157] In a preferred embodiment, the immunogenicity of the protein variants is determined 
experimentally to confirm that the variants do have reduced or eliminated immunogenicity 
relative to the parent protein. Several methods can be used for experimental confirmation of 
epitopes. In a preferred embodiment, ex vivo T-cell activation assays are used to 
experimentally quantitate immunogenicity. In this method, antigen presenting celts and 
naive T cells from matched donors are challenged with a peptide or whole protein of interest 
one or more times. Then, T cell activation can be detected using a number of methods, for 
example by monitoring production of cytokines or measuring uptake of tritiated thymidine. In 
the most preferred embodiment, interferon gamma production is monitored using Elispot 
assays (Schmittel et al, 2000, J. Immunol. Meth., 24: 17-24). If sera are available from 
patients who have raised an immune response to protein, it is possible to detect mature T 
cells that respond to specific epitopes. In a preferred embodiment, interferon gamma or IL-5 
production by activated T-cells is monitored using Elispot assays, although it is also possible 
to use other indicators of T cell activation or proliferation such as tritiated thymidine 
incorporation or production of other cytokines. Other suitable T cell assays include those 
disclosed in Meidenbauer ef a/., 2000, Prostate 43, 88-100; Schultes & Whiteside, 20O3, J. 
Immunol. Methods 279, 1-15; and Stickler et al., 200, J. Immunotherapy, 23, 654-660. In a 
preferred embodiment, the PBMC donors used for the above-described T cell activation 
assays will comprise class II MHC alleles that are common in patients requiring treatment for 
protein responsive disorders. For example, for most diseases and disorders, it is desirable 



40 



WO 2005/056759 



PCT7US2004/040694 



to test donors comprising all of the alleles that are prevalent in the population. However/for 
diseases or disorders that are linked with specific MHC alleles, it may be more appropriate to 
focus screening on alleles that confer susceptibility to protein responsive disorders. In a 
preferred embodiment, the MHC haplotype of PBMC donors or patients that raise an 
immune response to the wild type or protein variant are compared with the MHC haplotype 
of patients who do not raise a response. This data may be used to guide preclinical and 
clinical studies as well as aiding in identification of patients who will be especially likely to 
respond favorably or unfavorably to the protein therapeutic. 

[1 58] In an alternate preferred embodiment, immunogenicity is measured in transgenic 
mouse systems. For example, mice expressing fully or partially human class II MHC 
molecules may be used. In an alternate embodiment, immunogenicity is tested by 
administering the protein variants to one or more animals, including rodents and primates, 
and monitoring for antibody formation. Nonhuman primates with defined MHC haplotypes 
may be especially useful, as the sequences and hence peptide binding specificities of the 
MHC molecules in nonhuman primates may be very similar to the sequences and peptide 
binding specificities of humans. Similarly, genetically engineered mouse models expressing 
human MHC peptide-binding domains may be used (see for example Sonderstrup et al., 
1999, Immunol. Rev. 172: 335-343; and Forsthuber era/., 2001, J. Immunol. 167: 119-125). 
[1 59] The biological properties of the proteins of the present invention may be 
characterized in cell, tissue, and whole organism experiments. As is known in the art, drugs 
are often tested in animals, including but not limited to mice, rats, rabbits, dogs, cats, pigs, 
and monkeys, in order to measure a drug's efficacy for treatment against a disease or 
disease model, or to measure a drug's pharmacokinetics, toxicity, and other properties. The 
animals may be referred to as disease models. Therapeutics are often tested in mice, 
including but not limited to nude mice, SCID mice, xenograft mice, and transgenic mice 
(including knockins and knockouts). Such experimentation may provide meaningful data for 
determination of the potential of the protein to be used as a therapeutic. Any organism, 
preferably mammals, may be used for testing. For example because of their genetic 
similarity to humans, monkeys can be suitable therapeutic models, and thus may be used to 
test the efficacy, toxicity, pharmacokinetics, or other property of the proteins of the present 
invention. Tests of the in humans are ultimately required for approval as drugs, and thus of 
course these experiments are contemplated. Thus the proteins of the present invention may 
be tested in humans to determine their therapeutic efficacy, toxicity, immunogenicity, 
pharmacokinetics, and/or other clinical properties. 



41 



WO 2005/056759 



PCT/US2004/040694 



[160] In one embodiment of the present invention, a variant antibody for a host as 
compared to a parent antibody includes two or more amino acid substitutions derived from 
two or more natural antibodies. In this embodiment, a first resultant variant string in the 
variant antibody is rendered most homologous to a first natural antibody, a second resultant 
variant string in the variant antibody is rendered most homologous to the corresponding 
string in an second natural antibody, the substitutions are not in a CDR, and at least one 
resultant string is not a consensus of homologous natural sequences. In a preferred 
embodiment, the variant strings in the variant antibody do not include CDR residues. In a 
further preferred embodiment, the first and second natural antibodies are from different 
subfamilies. 

[161] In another embodiment of the present invention, a variant antibody for a host as 
compared to a parent antibody includes two or more amino acid substitutions derived from 
three or more natural antibodies. In this embodiment, a first resultant variant string in the 
variant antibody is rendered most homologous to a first natural antibody, a second resultant 
variant string in the variant antibody is rendered most homologous to the corresponding 
string in an second natural antibody, a third resultant variant string in the variant antibody is 
rendered most homologous to the corresponding string in an third natural antibody, the 
substitutions are not in a CDR, and at least one resultant string is not a consensus of 
homologous natural sequences. In an additional embodiment, the first, second and third 
natural antibodies are from different subfamilies. If a further additional embodiment, the 
variant antibody further comprises a fourth resultant variant string that is rendered most 
homologous to the corresponding string in a fourth natural antibody. If an additional 
embodiment, the variant antibody includes at least one substitution that is made at a position 
that is not surface exposed, the first, second and third natural antibodies are from different 
antibody groups, one of the substitutions is made at a position that is part of the VH/VL 
interface, and at least one amino acid substitution is not a back mutation. 

[162] In other embodiments, the first, second and third natural antibodies are from different 
antibody groups. 

[163] In other embodiments, the variant antibody includes at least one substitution that is 
not surface exposed. 

[164] In other embodiments, at least one of the substitutions is made at a position that is 
part of the VH/VL interface. 

[165] In other embodiments, the variant antibody includes at least one amino acid 
substitution is not a back mutation. 



42 



WO 2005/056759 



PC1YUS2004/040694 



[166] In one embodiment of the present invention, a variant antibody for a host as 
compared to a parent antibody includes a variant VH antibody region with host string content 
(HSC) greater than about 75%, and a framework region homogeneity (FRH) less than about 
60%, wherein the HSC and FRH are calculated with a window size of 9. 

[167] In one embodiment of the present invention, a variant antibody for a host as 
compared to a parent antibody includes a variant VH antibody region with exact string 
content greater than about 20%; and a framework region homogeneity less than about 60%, 
wherein the HSC and FRH are calculated with a window size of 9. 

[168] In one embodiment of the present invention, a variant antibody for a host as 
compared to a parent antibody includes a variant VL antibody region with exact string 
content greater than about 35%, and a framework region homogeneity less than about 60%, 
wherein HSC and FRH are calculated with a window size of 9. 

[1 69] In one embodiment of the present invention, a variant antibody for a host as 
compared to a parent antibody includes a first set of one or more amino acid substitutions 
from a first natural antibody and a second set of one or more amino acid substitutions from a 
second natural antibody, wherein the identity of said substituted amino acids from said 
second antibody differ from the corresponding amino acids of said first natural antibody, the 
substitutions are not in a CDR, and at least one substitution is not a consensus of 
homologous natural sequences. In an additional embodiment, the variant antibody further 
includes a third set of one or more amino acids substitutions from a third natural antibody 
wherein the identity of the substituted amino acids of said third set differ from the identity the 
corresponding amino acids from said first and second sets of amino acid substitutions. In a 
further embodiment, the variant antibody includes a fourth set of one or more amino acids 
from a fourth natural antibody wherein the identity of the substituted amino acids of said 
fourth set differ from the identity the corresponding amino acids from said first, second and 
third sets of amino acid substitutions. In other embodiments, the variant antibody includes 
multiple sets of one or more amino acids from multiple natural antibodies, wherein the 
identity of the substituted amino acids of any set drffer from the identity the corresponding 
amino acids from the other sets of amino acid substitutions. 

EXAMPLES 

[170] Examples are provided below to illustrate the present invention. These examples are 
not meant to constrain the present invention to any particular application or theory of 
operation. 



43 



WO 2005/056759 



PCTAJS2004/040694 



[171] For reference to immunoglobulin variable regions, positions are numbered according 
to the Kabat numbering scheme. For reference to immunoglobulin constant regions, 
positions are numbered according to the EU index as in Kabat (Kabat et a/., 1991 , 
Sequences of Proteins of Immunological Interest, 5th Ed., United States Public Health 
Service, National Institutes of Health, Bethesda). 

Example 1. Immunoqenicity Reduction of AC10 

[172] To illustrate application of the method described in the present invention, and to 
validate its broad applicability to immunogenicity reduction of proteins, a xenogeneic 
antibody example is provided using as the parent sequence the anti-CD30 antibody AC 10 
(Bowen et a/. Journal of Immunology, 1993, 151: 5896). A structural model of the mouse 
AC10 variable region was constructed using standard antibody modeling methods known in 
the art. Figures 3 and 4 show the sequences, host string content, and structures of the 
AC10 VL and VH domains (referred to as L0 AC10 VL and HO AC10 VH respectively). A 
CDR graft of this antibody was constructed by placing the AC 10 CDRs into the context of the 
frameworks of the most homologous host germlines, determined to be vlk_4-1 for VL and 
vhJ-3 for VH using the sequence alignment program BLAST. The sequences and string 
content of these CDR grafts are shown in Figures 5 and 6, along with structures of modeled 
AC10 highlighting the mutational differences between the CDR grafted AC10 variable chains 
and WT. 

[173] AC10 variants with reduced immunogenicity were generated by applying a string 
optimization algorithm on the WT AC1 0 VL and VH sequences. This algorithm heuristically 
samples multiple amino acid mutations that exist in the diversity of the human VUc and VH 
germline sequences, and calculates the host string content (HSC) of each sequence 
according to Equation 3 described above, using a window size w=9. In this set of 
calculations, residues in the CDRs and close to a CDR or to the VL/VH interface were 
masked, that is were not allowed to mutate. CDRs were defined as a slightly smaller set of 
residues than the CDRs defined by Chothia (Chothia & Lesk, 1987, J. Mol. Biol. 196: 901- 
917; Chothia et a/., 1989, Nature 342: 877-883; Al-Lazikani etal. f 1997, J. Mol Biol. 273: 
927-948). For the purposes of the present invention, VL CDRs are herein defined to include 
residues at positions 27-32 (CDR1), 50-56 (CDR2), and 91-97 (CDR3), wherein the 
numbering is according to Chothia. Because the VL CDRs as defined by Chothia and Kabat 
are identical, the numbering of these VL CDR positions is also according to Kabat. For the 
purposes of the present invention, VH CDRs are herein defined to include residues at 
positions 27-33 (CDR1), 52-56 (CDR2), and 95-102 (CDR3), wherein the numbering is 



44 



WO 2005/056759 



PCTAJS2004/040694 



according to Chothia. These VH CDR positions correspond to Kabat positions 27-35 
(CDR1), 52-56 (CDR2), and 95-102 (CDR3). Masked residues in these calculations were 
set at positions 1-4, 25-34, 36, 38, 43, 44, 46, 48-58, 60, 63-69, 71 , 87, and 89-98 for VL, 
and 2, 4, 24, 26-35, 37, 39, 44, 45, 47, 50-58, 60, 61, 71, 73, 76, 78, 91, and 93-106 for VH, 
wherein the numbering is according to Kabat Masking of potentially critical residues is a 
conservative approach to generating more host antibody variants, however it is but one 
embodiment of the present invention, and calculations wherein positions are not masked are 
also contemplated. This calculation was run for AC10 VL and VH in 100 separate 
interactions, generating a set of diverse AC 10 variants with more host string content than 
WT. Figure 7 shows the clustered nonredundant set of output sequences from these 
calculations for the AC10 VL and VH region, referred to as AC10 VL HSC Calculation 1 and 
AC10 VH HSC Calculation 1 respecitvely. For each iteration (Iter), the HSC (Eqation 3), 
HSS (Equation 5), and number (Mut) and identity (shaded residues) of mutations from WT 
are presented. In addition to the HSC score, each sequence was evaluated for its structural 
and functional integrity using a nearest neighbor structure-based scoring method (USSN 
60/528,229, filed December 8, 2003, entitled Protein Engineering with Analogous Contact 
Environments). Two measures of structural fitness, referred to as "Structural Consensus" 
and "Structural Precedence", are also provided in the Figure 7. Although the Analogous 
Contact Environments method is particularly well-suited for antibodies because of the wealth 
of sequence and structure information, any structure-based and/or sequence-based scoring 
method may be used to evaluate the structural and functional fitness of the variant 
sequences. The output sequences were clustered based on their mutational distance from 
the other sequences in the set, and these clusters are delineated by the horizontal black 
lines in the Figure. The "Cluster" column provides the quantitative mutational distance 
between each sequence and the rest of the sequence in its cluster; sequences with a lower 
cluster value are more representative of that particular sequence cluster. 

[174] These calculations were used to generate a set of AC10 VL and VH variants. In 
some cases, further substitutions were made to output sequences, using string and 
structural scores, as well as visual inspection of the modeled AC10 structure, to evaluate 
fitness. Figures 8-13 present the sequences, host string content, and mapped mutational 
differences on the modeled AC 10 structure for each of the AC 10 VL and VH variants. 
Iteration 36 from AC10 VL HSC calculation 1 served as the precursor for L1 AC10 VL, 
iteration 37 served as the precursor for L2 AC 10 VL, and iteration 3 served as the precursor 
for L3 AC10 VL. Iteration 15 from AC10 VH HSC calculation 1 served as the precursor for 
H1 AC 10 HL, iteration 55 served as the precursor for H2 AC 10 VH, and iteration 18 served 



45 



WO 2005/056759 



PCTAJS2004/040694 



as the precursor for H3 AC10 VH. 

[175] Tables 1 and 2 present the number of mutations from the parent sequence, structural 
fitness scores, and host string scores for the AC10 VL and VH variants as compared to the 
WT and CDR grafted AC10 sequences. In addition to the aforementioned structural and 
host string analysis, each sequence was analyzed for its global homology to the host 
germline. The maximum identity match to the germline for each string in the sequences was 
also determined, referred to as N| D max. This represents the total number of strings in each 
sequence whose maximum identity to the corresponding strings in the host germline is the 
indicated value. For w = 9, Tables 1 and 2 list N 9 max, N 8 max, N 7 max, and N^max for each 
sequence. N 9 max represents the number of strings in the sequence for which 9 of 9 
residues match at least one string in the host germline, N 8 max represents the number of 
strings for which 8 of 9 residues match at least one string in the host germline, N 7 max 
represents the number of strings for which 7 of 9 residues match at least one string in the 
host germline, and N^max represents the number of strings for which 6 or less residues of 9 
residues match at least one string in the host germline. This last category (ID <6) could, for 
example, be regarded as the number of poorly scoring strings. In addition to the 
aforementioned structural and host string analysis, each sequence was analyzed for its 
global homology to the host germline; Tables 1 and 2 present the most homologous human 
germline sequence for each sequence (Closest Germline) and corresponding identity to that 
germline (ID to Closest Germline), determined using the sequence alignment program 
BLAST. Finally, the Framework region homogeneity (FRH) of each variant was evaluated 
for w=9, and is presented in Tables 1 and 2, providing the extent to which the host string 
content of each variant is derived from a single germline as opposed to multiple germline 
sequences. 

Table 1. AC10 VL Variants 

L1 L2 L3 

15 16 9 
0.59 0.64 0.56 

0.66 0.67 0.58 

0.86 0.86 0.85 

0.48 0.46 0.41 



WT CDR 

Wl Graft 

Mutations 18 

Structural 



Consensus 

Structural 
Precedence 

Human String 
Content 

Human String 
Similarity 



0.57 0.57 

0.68 0.57 

0.78 0.88 

0.15 0.57 



46 



WO 2005/056759 PCT/US2004/040694 



WT 



CDR 
Graft 



Framework Region 
Homogeneity 

N 9 max 15 61 

N 8 max 27 11 

N 7 max 31 15 

N^max 34 20 



4-1 4-1 



L1 


L2 


L3 


0.73 


0.81 


0.52 


51 


48 


42 


13 


15 


21 


21 


22 


20 


22 


22 


24 


3-11 


1-39 


4-1 



Closest 
Germline 

ID to Closest 68/101 867 101 78 / 99 80 / 99 75/101 

Germline 67% 85% 79% 81% 74% 



Table 2. AC10 VH Variants 





WT 


CDR 
Graft 


H1 


H2 


H3 


Mutations 




26 


16 


23 


20 


Structural 


0.49 


0.48 


0.48 






Consensus 


0.50 


0.47 


Structural 


0.63 


0.67 


0.63 






Precedence 


0.59 


0.59 


Human String 


0.69 


0.87 


0.81 






Content 


0.81 


0.80 


Human String 
Similarity 


0.07 


0.68 


0.41 


0.39 


0.38 


Framework 












Region 


0.60 


0.86 


0.65 


0.47 


0.55 


Homogeneity 












N 9 max 


5 


81 


48 


45 


44 


N 8 max 


31 


13 


30 


32 


28 


N 7 max 


34 


8 


20 


21 


24 


N^max 


49 


17 


21 


21 


23 


Closest 


1-3 


1-3 


1-3 






Germline 


1-3 


7-4-1 


ID to Closest 


69 / 98 93 / 98 83 198 721 98 76 / 9£ 


Germline 


70% 


95% 


85% 


73% 


78% 



[176] An important observation is that, whereas the CDR grafted antibodies are most 
homologous to a single human germline sequence (the "acceptor" sequence in humanization 
terminology), the present invention describes variants that are homologous to different host 
germline sequences in different regions of the sequence. This is evident from the significant 
differences in Framework region homogeneity (FRH) scores for the AC 10 variants of the 
present invention and CDR grafted AC10 variants. Furthermore, whereas CDR grafted 



47 



WO 2005/056759 



PCTAJS2004/040694 



AC10 VL and VH are most homologous to human germline subfamilies 4 (VL) and 1 (VH) 
respectively across their entire sequences, a number of the AC10 variants are most 
homologous to different subfamilies in different frameworks. Additionally, whereas the CDR 
grafted antibodies are most homologous to a single germline sequence that is also the most 
homologous sequence to the parent sequence, the present invention presents a set of 
antibodies for a given antibody that are most homologous to different human germline 
sequences, which need not be the most homologous germline sequence to WT. For 
example, Table 1 shows that CDR grafted AC10 VL is most homologous to 4-1 , which is 
also the most homologous human germline to the WT AC10 parent. However L1, L2, and 
L3 are most homologous to three different human germlines -3-11, 1-39, and 4-1 
respectively. Thus the variants of the present invention explore a substantially greater 
amount of diversity than CDR grafted antibodies. One obvious advantage of this is that the 
method of the present invention provides a greater chance of success with respect to 
antigen affinity. The choice of an "acceptor" in humanization methods places a single bet; if 
the donor CDRs are in fact incompatible with the acceptor FRs, a set of backmutations that 
regain WT affinity may not exist. In contrast, the method of the present invention enables a 
greater diversity of sequence and structure space to be sampled in the immunogenicity 
reduction process, increasing the chances of obtaining a final less immunogenic version with 
WT affinity or better. An additional advantage of sampling greater sequence diversity is that 
some sequences may have more optimal properties than others, for example with regard to 
stability, solubility, and effector function. For example, as disclosed in USSN 60/614,944, 
and USSN 60/619,409, filed October 14, 2004, entitled "Immunoglobulin Variants Outside 
the Fc Region with Optimized Effector Function", the variable region of an antibody may 
impact effector functions such as antibody dependent cell-mediated cytotoxicity (ADCC), 
antibody dependent cell-mediated phagocytosis (ADCP), and complement dependent 
cytotoxicity (CDC). 

[177] The genes for the variable regions of AC1 0 WT (L0 and HO) and variants (L1 , L2, L3, 
H1, H2, and H3) were constructed using recursive PCR, and subcloned into a the 
mammalian expression vector pcDNA3.1Zeo (Invitrogen) comprising the full length light 
kappa (CU) and heavy chain lgG1 constant regions. All sequences were sequenced to 
confirm the fidelity of the sequence. Plasmids containing heavy chain gene (VH-CH1-CH2- 
CH3) (wild-type or variants) were co-transfected with plasmid containing light chain gene 
(VL-CLO) in all combinations (L0/H0, L0/H1, L0/H2, L0/H3, L1/H0, L1/H1, L1/H2, L1/H3, 
L2/H0, L2/H1 , L27H2, L2/H3, L3/H0, L3/H1 , L3/H2, L3/H3) into 293T cells. Here, for 
example, L2/H3 refers to the L2 AC10 VL paired with H3 AC10 VH. Media were harvested 5 



48 



WO 2005/056759 



PCT/US2004/040694 



days after transfection, and antibodies were purified from the supernatant using protein A 
affinity chromatography (Pierce, Catalog # 20334). 

[178] WT and variant antibodies were experimentally tested for their capacity to bind CD30 
antigen. Binding affinity to human CD30 by the AC10 WT and variant antibodies was 
measured using a quantitative and extremely sensitive method, AlphaScreen™ assay. The 
AlphaScreen™ assay is a bead-based non-radioactive luminescent proximity assay. Laser 
excitation of a donor bead excites oxygen, which if sufficiently close to the acceptor bead will 
generate a cascade of chemiluminescent events, ultimately leading to fluorescence emission 
at 520-620 nm. The AlphaScreen™ assay was applied as a competition assay for screening 
the antibodies. WT AC10 antibody was biotinylated by standard methods for attachment to 
streptavidin donor beads (Perkin Elmer). Commericial CD30 was conjugated to digoxigenin 
(DIG) (Roche Diagnostics) for attachment to anti-DIG acceptor beads (Perkin Elmer). In the 
absence of competing AC10 variants, WT antibody and CD30 interact and produce a signal 
at 520-620 nm. Addition of untagged ACiO variant competes with the WT AC10 / CD30 
interaction, reducing fluorescence quantitatively to enable determination of relative binding 
affinities. Figures 14a and 14b show binding of WT (H0L0) and AC10 variant antibodies to 
CD30 using the AlphaScreen™ assay. The data were fit to a one site competition model 
using nonlinear regression, and these fits are represented by the curves in the figure. These 
fits provide the inhibitory concentration 50% (IC50) (i.e. the concentration required for 50% 
inhibition) for each antibody, thus enabling the relative binding affinities relative to WT to be 
determined. Table 3 provides the IC50's and Fold lC50's relative to WT for fits to these 
binding curves. The AC10 variants display an array of CD30 binding affinities, with a 
number of variants binding CD30 with affinity comparable to or better affinity than WT AC10. 

[179] Antigen affinity of the AC10 variants was also measured using Surface Plasmon 
Resonance (SPR) (Biacore, Uppsala, Sweden). SPR allows for the measurement of direct 
binding rates and affinities of protein-protein interactions, and thus provides an excellent 
complementary binding assay to the AlphaScreen™ assay. CD30 fused to the Fc region of 
lgG1 (R&D Systems) was immobilized on a Protein A SPR chip, the surface was blocked 
with Fc, and WT and variant AC 10 antibodies were flowed over the chip at a range of 
concentrations. The resulting sensorgrams are shown in Figure 15. Global Langmuirfrts 
were carried out for the concentrations series using the BiaEvaluation curve fitting software, 
providing the on-rate constant (ka), off-rate constant (kd), and equilibrium binding constant 
(KD=kd/ka) for the curves. Table 3 provides the KDs and Fold KDs relative to WT for the 
SPR data. The excellent agreement between the rank ordering of the variants as 
determined by SPR and AlphaScreen™ assay support the accuracy of the data. 



49 



WO 2005/056759 



PCMJS2004/040694 



Table 3. CD30 Binding of AC10 Variants 



AC10 
Variant 


SPR 
KD (nM) 


SPR 
Fold KD 


AlphaScreen 
IC50(nM) 


AlphaScreen 
Fold IC50 


H2L1 


9.49 


0.36 


55.1 


0.06 


H2L2 


5.95 


0.57 


49.2 


0.06 


H1L2 


7.55 


0.45 


45.3 


0.07 


H1L1 


5.63 


0.60 


27.7 


0.11 


H2L3 


6.75 


0.50 


27.2 


0.12 


H2L0 


8.00 


0.42 


19.4 


0.16 


H1L3 


5.09 


0.67 


17.4 


0.18 


H1L0 


6.39 


0.53 


9.77 


0.32 


H0L2 


3.48 


0.97 


7.81 


0.41 


H3L2 


2.86 


1.19 


6.57 


0.48 


H3L0 


3.08 


1.10 


6.18 


0.51 


H3L1 


2.44 


1.39 


6.09 


0.52 


H0L1 


3.29 


1.03 


5.19 


0.61 


H0L3 


3.00 


1.13 


4.61 


0.69 


H0LO 


3.39 


1.00 


3.18 


1.00 


H3L3 


2.33 


1.45 


1.99 


1.59 



[180] In addition to assessing the antigen affinity and biophysical properties of the variants 
of the present invention, they may also be tested for effector functions in the context of a full 
length antibody. One advantage of generating multiple reduced immunogenicity variants of 
a parent immunoglobulin is that it enables a greater degree of sequence diversity to be 
sampled, diversity which may provide optimal properties. Some sequences may have more 
optimal properties than others, for example with regard to effector function. For example, as 
disclosed in USSN 60/614,944, and USSN 60/619,409, filed October 14, 2004, entitled 
"Immunoglobulin Variants Outside the Fc Region with Optimized Effector Function", the 
variable region of an antibody may impact effector functions such as antibody dependent 
cell-mediated cytotoxicity (ADCC), antibody dependent cell-mediated phagocytosis (ADCP), 
and complement dependent cytotoxicity (CDC). 

[181] In order to explore any differences in capacity to mediate effector function, the 
affinities of the AC10 variants for FcKRIIIa were measured using the AlphaScreen™ assay. 
The extracellular region of human V158 F^RHIa was obtained by PCR from a clone 
obtained from the Mammalian Gene Collection (MGC:22630), and the receptor was fused 
with glutathione S-Transferase (GST) to enable screening. Tagged FcKRIIIa was 
transfected in 293T cells, and media containing secreted FcKRIIIa were harvested and 
purified. The AlphaScreen™ assay was applied as a competition assay for screening AC10 
variants for binding to FcKRIIIa. Biotinylated WT AC10 antibody was bound to streptavidin 



50 



WO 2005/056759 



PCT/US2004/040694 



donor beads (Perkin Elmer), and GST-fused human V158 F^RIHa was bound to anti-GST 
acceptor beads (Perkin Elmer). The binding data are shown in Figures 16a and 16b, and 
the resulting IC50's and Fold IC50's relative to WT are provided in Table 4. FcKRIIIa affinity 
of the AC10 variants was also measured using SPR. GST-fused human FcKRIIIa (V158 
isoform) was immobilized on a chip, and WT and variant AC10 antibodies were flowed over 
the chip at a range of concentrations. Binding constants were obtained from fitting the data 
using standard curve-fitting methods, The equilibrium dissociation constants (KDs) obtained 
from the fits to these binding curves, and the calculated fold improvement or reduction 
relative to WT (Fold KD) are shown in Table 4. 



Table 4. FcKRIIIa Binding of AC10 Variants 



AC10 
Variant 


SPR 
KD {nM) 


SPR 
Fold KD 


AlphaScreen 
IC50 (nM) 


AlphaScreen 
Fold IC50 


H2L1 


14.9 


1.25 


751 


0.12 


H2L2 


4.01 


4.64 


146 


0.60 


H1L2 


1.6.6 


1.12 


340 


0.26 


H1L1 


11.2 


1.66 


221 


0.39 


H2L3 


3.52 


5.28 


183 


0.48 


H2L0 


12.9 


1.44 


175 


0.50 


H1L3 


11.2 


1.66 


178 


0.49 


H1L0 


22.0 


0.85 


71.6 


1.22 


H0L2 


9.09 


2.05 


93.8 


0.93 


H3L2 


3.57 


5.21 


88.7 


0.98 


H3L0 


20.0 


0.93 


216 


0.40 


H3L1 


17.4 


1.07 


209 


0.42 


H0L1 


11.6 


1.60 


183 


0.48 


H0L3 


12.7 j 


1.46 


146 


0.60 


H0L0 


18.6 


1.00 


87.2 


1.00 


H3L3 


6.13 


3.03 


83.5 


1.04 



[182] To assess the capacity of the AC10 variants to mediate effector function against 
CD30 expressing cells, the AC 10 variants were tested in a cell-based ADCC assay. Human 
peripheral blood monocytes (PBMCs) were isolated from buffy-coat and used as effector 
cells, and CD30 positive L540 Hodgkin's lymphoma cells were used as target cells. L540 
target cells were seeded at 20,000 per well in 96-welI plates and treated with designated 
antibodies in triplicates starting at 1 pg/ml and in reduced concentrations in 34 log steps. 
PBMCs isolated using a Ficoll gradient and allotyped as FcKRIIIa 158 V/F were added at 25- 
fold excess of L540 cells and co-cultured for 4 hrs before processing for LDH activity using 
the Cytotoxicity Detection Kit (LDH, Roche Diagnostic Corporation, Indianapolis, IN) 
according to the manufacturer's instructions. The plates were read using a Wallac 1420 



51 



WO 2005/056759 



PCTAJS2004/040694 



Victor 2 ™. Figures 17a - 17c show the results. The graphs show that the antibodies differ 
not only in their EC50, reflecting their relative potency, but also in the maximal level of ADCC 
attainable by the antibodies at saturating concentrations, reflecting their relative efficacy. 
These two terms, potency and efficacy, are sometimes used loosely to refer to desired 
clinical properties. In the current experimental context, however, they are denoted as 
specific quantities, and therefore are here explicitly defined. By " potency" as used in the 
current experimental context is meant the EC50 of an EGFR targeting protein. By " efficacy" 
as used in the current experimental context is meant the maximal possible effector function 
of an antibody at saturating levels. Differences in capacity to mediate ADCC may be due to 
differences in antigen affinity, different capacities of the variant variable regions to effect 
FckR binding, or both. Regardless, the contribution of an antibody variable region to Fc/R 
binding and effector function may be an important parameter for selecting a clinical 
candidate. The choice of an antibody clinical candidate based in whole or in part on the 
impact on effector function of the variable region represents a novel dimension in antibody 
therapeutics. 

[183] Based on the CD30 binding, FcKRIIIa binding, and ADCC results, the H3/L3 AC10 
variant was chosen as a potential biotherapeutic candidate. Because this antibody is 
intended for clinical use as an anti-cancer therapeutic, it may be advantageous to optimize 
its effector function. As previously described, substitutions can be engineered in the 
constant region of an antibody to provide favorable clinical properties. In a most preferred 
embodiment, one or more amino acid modifications that provide optimized binding to Fc/Rs 
and/or enhanced effector function described in USSN 10/672,280, PCT US03/30249, and 
USSN 10/822,231, and USSN 60/627,774, filed 11/12/2004 and entitled "Optimized Fc 
Variants", are combined with the AC10 variants of the present invention. A number of 
optimized Fc variants obtained from these studies, including I332E, S239D, V264I/I332E, 
S239D/I332E, and S239D/A330L/I332E, were constructed in the H0/L0 and H3/L0 AC10 
antibodies using quick change mutagenesis (Stratagene). Antibodies were expressed and 
purified as described above. Figures 18a and 18b show the results of the ADCC assay, 
carried out as described above, comparing WT (H0/L0) and H3/L3 AC10 in combination with 
the optimized Fc variants. Considerable enhancements in potency and efficacy are 
observed for the Fc variant antibodies as compared to H0/L0 and H3/L3 AC 10. 

[184] As described above, because variant sequences of the invention are preferably 
derived from a HSC-increasing procedure in which substitution of structurally important 
positions is disallowed (or discouraged), it is likely that additional optimization of HSC is 
possible if those positions are allowed to vary in a secondary analysis. It is noted that, due 



52 



WO 2005/056759 



PCTAJS2004/040694 



to residue masking, mutations in the variants occur distal to the CDRs and VL/VH interface. 
This is in contrast to CDR grafted antibodies, which have mutations in the parent that are at 



antigen affinity. This is corroborated by the fact that CDR grafted antibodies typically require 
backmutations to the donor sequence to regain WT affinity for antigen. Such backmutations 
are usually made out of structural and immunogenic context with respect to host sequences, 
and cause dramatic reductions in the host string content of the final variant. In contrast, the 
variants presented herein are simultaneously optimized for host string and structural fitness 
within the same context, and no backmutations need be made. Nonetheless, one or more 
subsequent substitutions may be explored to increase antigen affinity or further improve 
HSC, for example by mutating residues that were masked in the calculations and/or residues 
in or close to the CDRs or VL/VH interface. Thus the H3/L3 variant can be thought of as a 
primary variant or template for further optimization, and variants of H3/L3 can be thought of 
as secondary variants. In contrast to backmutating as with CDR grafted antibodies, 
secondary substitutions in the variants of the present invention will comprise forward or 
neutral mutations with respect to the host germline, and thus are expected to only improve or 
unaffected HSC. An additional benefit of generating secondary variants is that, by exploring 
quality structural and string diversity, it is also possible that other properties can be 
optimized, for example affinity, activity, specificity, solubility, expression level, and effector 



[185] String analysis was carried out on the H3/L3 sequence to design a set of secondary 
substitutions that have neutral, positive, or minimal impact on HSC, and/or that have 
significant potential for optimization of antigen affinity and/or effector function. Table 5 
provides this set of 70 VL (Table 5a) and 64 VH (Table 5b) single mutations. The H3 column 
provides the WT H3 amino acid, and the Sub column provides the designed substitution. 
Positions are numbered according to the Kabat numbering format, with Kabat CDR positions 
bolded. The provided string impact defined according to Equation 7, describes the 
difference in HSC between the primary variant sequence, here H3/L3, and the secondary 
variant sequence. 



or near these critical regions and thus have a significantly greater potential for perturbing 



function. 



Table 5a. L3 AC10 Secondary Variants 



Variant 



Pos 
(Kabat) 



L3 Sub 



String Fold Fold Fold 
Impact ProtA CD30 FcrRMIa 



L3.1 
L3.2 



1 
1 



D 
D 



A 
E 



0 
1 



0.89 
1.09 



1.30 
1.24 



0.96 
1.46 



53 



WO 2005/056759 



Variant 



L3.3 

L3.4 

L3.5 

L3.6 

L3.7 

L3.8 

L3.9 

L3.10 

L3.11 

L3.12 

L3.13 

L3.14 

L3.15 

L3.16 

L3.17 

L3.18 

L3.19 

L3.20 

L3.21 

L3.22 

L3.23 

L3.24 

L3.25 

L3.26 

L3.27 

L3.28 

L3.29 

L3.30 

L3.31 

L3.32 

L3.33 

L3.34 

L3.35 

L3.36 

L3.37 

L3.38 

L3.39 

L3.40 

L3.41 

L3.42 

L3.43 

L3.44 

L3.45 

L3.46 

L3.47 

L3.48 

L3.49 

L3.50 



PCT/US2004/040694 



Pos 


L3 


Sub 


String 


Fold 


Fold 


Fold 


(Kabat) 






Impact 


ProtA 


CD30 


FcKRIIIa 


1 


D 


N 


0 


1.24 


1.66 


1.35 


1 


D 


S 


0 


0.97 


1.04 


1.18 


3 


V 


Q 


0 


1.13 


1.32 


1.32 


4 


L 


M 


.4 


1.65 


1.64 


1.21 


25 


A 


S 


6 








27a 


S 


D 


0 


1.02 


0.94 


0.61 


27b 


V 


I 


1 


1.04 


0.58 


0.81 


27c 


D 


L 


5 








27c 


D 


S 


8 








27c 


D 


V 


3 


1.24 


1.15 


1.19 


27d 


F 


D 


0 


1.07 


0.32 


0.98 


27d 


F 


H 


4 


0.86 


0.08 


0.93 


27d 


F 


Y 


5 


i.04 


0.63 


1.19 


28 


D 


N 


-1 


1.10 


1.61 


1.18 


30 


D 


K 


4 


1.01 


1.21 


1.24 


30 


D 


N 


6 


1.09 


1.45 


0.94 


30 


D 


S 


4 


1.07 


1.13 


0.82 


30 


D 


Y 


0 


0.82 


0.78 


0.73 


31 


S 


D 


0 


1.01 


0.81 


0.95 


31 


S 


T 


3 


1.03 


0.46 


0.97 


31 


S 


N 


-1 


1.03 


0.71 


1.00 


32 


Y 


D 


0 


1.31 


0.46 


1.33 


33 


M 


L 


8 


1.38 


1.36 


1.37 


34 


N 


S 


0 








34 


N 


A 


-1 


1.39 


0.38 


1.36 


34 


N 


D 


-6 


1.19 


0.41 


1.76 


46 


V 


H 


4 


0.06 


0.03 


0.11 


46 


V 


L 


9 


0.86 


0.39 


0.75 


46 


V 


R 


4 








46 


V 


S 


4 


1.05 


0.32 


0.90 


50 


A 


D 


6 


0.98 


0.26 


0.66 


50 


A 


S 


5 


1.01 


0.47 


1.20 


50 


A 


W 


2 








53 


N 


s 


8 








53 


N 


T 


5 


0.99 


1.26 


1.01 


54 


L 


R 


1 


1.19 


1.46 


1.55 


55 


E 


A 


2 


1.01 


0.85 


1.32 


55 


E 


Q 


6 


0.99 


0.87 


1.07 


56 


S 


T 


8 


1.50 


1.80 


1.23 


58 


I 


V 


4 


1.44 


1.55 


0.95 


60 


A 


D 


1 


1.11 


1.16 


1.08 


60 


A 


S 


2 


0.82 


1.08 


0.85 


67 


S 


P 


0 








89 


Q 


H 


1 


1.37 


0.08 


1.64 


91 


S 


A 


8 








91 


S 


G 


9 


0.85 


0.29 


0.80 


91 


S 


H 


2 


.1.20 


0.01 


1.32 


91 


S 


L 


8 


1.10 


0.02 


1.59 



54 



WO 2005/056759 



PCT/US2004/040694 



Variant 



Pos 
(Kabat) 



L3 



Sub , Strin 9 
Impact 



Fold 
Prot A 



Fold 
CD30 



Fold 
FcKRIHa 



L3.51 


91 


s 


Y 


8 


1.00 


0.02 


1.50 


L3.52 


92 


N 


I 


3 








L3.53 


92 


N 


S 


2 


3.02 


0.48 


1.34 


L3.54 


92 


N 


Y 


8 


1.39 


0.96 


1.05 


L3.55 


93 


E 


K 


8 


0.62 


0.27 


0.49 


L3.56 


93 


' E 


N 


8 


1.06 


0,64 


0.84 


L3.57 


93 


E 


Q 


2 








L3.58 


93 


E 


S 


8 


0.90 


0.49 


0.87 


L3.59 


94 


D 


A 


3 


1.16 


0.09 


1.14 


L3.60 


94 


D 


F 


9 


1.22 


0.02 


1.19 


L3.61 


94 


D 


H 


8 








L3.62 


94 


D 


L 


3 


0.87 


0.46 


0.79 


L3.63 


94 


D 


S 


1 


1.74 


0.57 


1.42 


L3.64 


94 


D 


T 


7 


1.24 


0.14 


1.16 


L3.65 


96 


W 


F 




0.33 


0.34 


0.29 


L3.66 


96 


W 


I 




0.75 


0.00 


0.57 


L3.67 


96 


w 


L 










L3.68 


96 


w 


Y 










L3.69 


100 


G 


P 










L3.70 


100 


G 


Q 











Table 5b. H3 AC10 Secondary Variants 



Variant 


Position 
(Kabat) 


H3 


Sub 


String 
Impact 


Fold 
Prot A 


Fold 
CD30 


H3.1 


1 


Q 


E 


-1 


0.83 


1.00 


H3.2 


2 


I 


L 


0 


1.60 


2.76 


H3.3 


2 


I 


M 


2 


0.88 


0.68 


H3.4 


2 


I 


V 


0 


0.98 


1.28 


H3.5 


9 


P 


A 


2 


0.95 


1.29 


H3.6 


16 


A 


T 


2 


0.89 


1.13 


H3.7 


24 


A 


V 


2 


1.54 


4.45 


H3.8 


31 


D 


G 


2 


0.80 


1.40 


H3.9 


31 


D 


S 


2 


0.82 


1.65 


H3.10 


33 


Y 


D 


2 


0.68 


0.07 


H3.11 


33 


Y 


G 


3 


0.96 


0.73 


H3.12 


33 


Y 


W 


0 


0.84 


0.00 


H3.13 


34 


I 


L 


1 


0.96 


1.52 


H3.14 


34 


I 


M 


8 


1.05 


1.62 


H3.15 


35 


T 


D 


1 


1.55 


0.05 


H3.16 


35 


T 


G 


2 


1.03 


0.15 


H3.17 


35 


T 


H 


8 


0.86 


0.04 


H3.18 


35 


T 


N 


4 


1.07 


0.13 


H3.19 


35 


T 


S 


6 


0.88 


1.11 


H3.20 


44 


G 


A 


0 


1.20 


2.04 


H3.21 


44 


G 


R 


0 


1.36 


2.60 


H3.22 


50 


W 


I 


6 


1.25 


0.01 



55 



WO 2005/056759 



PCTAJS2004/040694 



Variant 


Position 


H3 


Sub 


String 


Fold 


Fold 


(Kabat) 


impact 


ProtA 


CD30 


H3.23 


50 


W 


R 


0 


0.99 


0.16 


H3.24 


52 


Y 


N 


2 


1.03 


0.03 


H3.25 


52 


Y 


T 


•j 


1.11 


0.06 


H3.26 


52 


Y 


V 


•j 


1.33 


0.06 


H3.27 


52a 


P 


A 




1.02 


2.00 


H3.28 


52a 


P 


V 


•j 


1.44 


1.34 


H3.29 


54 


S 


D 




1.45 


1.81 


H3.30 


54 


S 


N 


5 


1.13 


1.45 


H3.31 


58 


K 


G 


4 






H3.32 


58 


K 


I 


2 


1.22 


1.09 


H3.33 


58 


K 


N 


5 


1.26 


0.50 


H3.34 


60 


N 


A 


7 


0.87 


1 30 


H3.35 


60 


N 


P 


o 






H3.36 


60 


N 


S 


7 


1.02 


1.24 


H3.37 


60 


N 


T 


0 


1.12 


1.01 


H3.38 


60 


N 


V 


0 


1.16 


1.14 


H3.39 


60 


N 


D 


0 


1.09 


1.00 


H3.40 


61 


E 


Q 


7 


1.51 


1.83 


H3.41 


64 


Q 


T 


0 


0.98 


1.38 


H3.42 


71 


V 


L 


4 


1.10 


0.66 


H3.43 


71 


V 


M 


9 


1.17 


0.88 


H3.44 


71 


V 


R 


1 


1.25 


1.76 


H3.45 


87 


T 


M 


-1 


0.99 


1.14 


H3.46 


89 


V 


M 


-4 


1.41 


1.39 


H3.47 


91 


F 


H 


2 






H3.48 


91 


F 


Y 


9 


1.32 


1.60 


H3.49 


93 


A 


T 


0 


1.47 


0.38 


H3.50 


93 


A 


V 


0 


1.01 


1 40 


H3.51 


94 


N 


A 


9 


1.51 


0.08 


H3.52 


94 


N 


H 


5 


1.23 


1.24 


H3.53 


94 


N 


K 


9 


1.67 


0 02 


H3.54 


94 


N 


R 


9 


1.26 


0.00 


H3.55 


94 


N 


T 


g 


1.24 


0 91 


H3.56 


99 


W 


Y 




1.26 


0.07 


H3.57 


101 


A 


D 




1.31 


0.53 


H3.58 


101 


A 


Q 




1.17 


0.16 


H3.59 


102 


Y 


H 




1.69 


1.05 


H3.60 


102 


Y 


S 




1.04 


0.82 


H3.61 


102 


Y 


V 




1.33 


1.21 


H3.62 


102 


Y 


L 




1.34 


1.22 


H3.63 


102 


Y 


F 




1.18 


1.24 


H3.64 


105 


Q 


R 




1.15 


1.28 



[186] The secondary. H3/L3 variants were constructed using quick change mutagenesis, 
and the full length antibodies were expressed and purified as described above. H3 variants 
comprised H3 variant VH chains(H3.1- H3.64) in combination with L3 VL, and L3 variants 
comprised L3 variant VL chains (L3.1 ~ L3.70) in combination with H3 VH. The 



56 



WO 2005/056759 



PO7US2004/040694 



AlphaScreen™ assay was used to measure binding of the H3/L3 secondary variants to 
CD30 and FcKRIHa (as described earlier), as well as to protein A using biotinylated AC 10 
bound directly to protein A acceptor beads and streptavidin donor beads. Figure 19 provides 
AlphaScreen™ binding curves for binding of select AC10 variants to CD30. The Fold IC50's 
relative to WT H3/L3 for binding to CD30, FcKRIHa, and protein A are provided in Table 5. A 
number of H3/L3 secondary variants provide comparable or improved binding to CD30 
antigen relative to the H3/L3 parent, enabling the engineering of additional variants that 
comprise combinations of these substitutions, which may provide further enhancements in 
HSC and/or antigen affinity. 

[187] Secondary substitutions that show favorable properties with respect to antigen 
affinity, effector function, stability, solubility, expression, and the like, may be combined in 
subsequent variants to generate a more optimized therapeutic candidate. Two new VL and 
three new VH variants were designed that comprise combinations of the described 
secondary substitutions, referred to as L3.71, L3 .72, H3.68, H3.69, and H3.70. Figures 20 - 
24 present the sequences, host string content, and mapped mutational differences on the 
modeled AC10 structure for each of these new AC10 VL and VH variants. Table 6 presents 
the number of mutations from the parent sequence, structural fitness scores, host string 
scores, and homology scores for these AC 10 VL and VH variants. 

Table 6. AC10 Variants 





L3.71 


L3.72 


H3.68 


H3.69 


H3.70 


Mutations 


15 


15 


23 


27 


30 


Structural 


0.56 


0.55 


0.46 






Consensus 


0.46 


0.45 


Structural 


0.54 


0.52 








Precedence 


0.55 


0.57 


0.56 


Human 












String 


0.88 


0.87 


0.80 


0.83 


0.84 


Content 












Human 












String 


0.52 


0.45 


0.39 


0.47 


0.47 


Similarity 












Framework 












Region 


0.47 


0.51 


0.33 


0.40 


0.42 


Homogeneity 












N 9 max 


55 


47 


46 


55 


55 


N 8 max 


19 


24 


26 


26 


34 


N 7 max 


14 


17 


24 


20 


12 


N^max 


19 


19 


23 


18 


18 



57 



WO 2005/056759 



PCTAJS2004/040694 





L3.71 


L3.72 


H3.68 


H3.69 


H3.70 


Closest 
Germline 


4-1 


4-1 


7-4-1 


1-3 


1-3 


ID to Closest 


76/101 


76/101 


74/98 


77/98 


79/98 


Germline 


75% 


75% 


76% 


79% 


81% 



[188] Because the provided AC 10 variants antibodies are clinical candidates for anti- 
cancer therapeutics, it may be advantageous to optimize their effector function. As 
previously described, substitutions can be engineered in the constant region of an antibody 
to provide favorable clinical properties. Combinations of the variants of the present invention 
with Fc modifications that alter effector function are anticipated. In a most preferred 
embodiment, one or more amino acid modifications that provide optimized binding to FcyRs 
and/or enhanced effector function described in USSN 10/672,280, PCT US03/30249, and 
USSN 10/822,231, and USSN 60/627,774, filed 11/12/2004 and entitled "Optimized Fc 
Variants", are combined with the AC10 variants of the present invention. The optimal antt- 
CD30 clinical candidate may comprise amino acid modifications that reduce immunogenicity 
and enhance effector function relative to a parent anti-CD30 antibody. Figures 25a - 25c 
provide the light and heavy chain sequences of AC10 variants that comprise L3.71/H3.70 
AC10 as described above, combined with a number of possible variant lgG1 constant 
regions, comprising one or more modifications at S239, V264, A330, and I332, that provide 
enhanced effector function. 

[189] Although human lgG1 is the most commonly used constant region for therapeutic 
antibodies, other embodiments may utilize constant regions or variants thereof of other IgG 
immunoglobulin chains. Effector functions such as ADCC, ADCP, CDC, and serum half-life 
differ significantly between the different classes of antibodies, including for example human 
lgG1, lgG2, lgG3, lgG4, lgA1, lgA2, IgD, IgE, IgG, and IgM (Michaelsen et al. t 1992, 
Molecular Immunology, 29(3): 319-326). A number of studies have explored lgG1, lgG2, 
lgG3, and lgG4 variants in order to investigate the determinants of the effector function 
differences between them. See for example Canfield & Morrison, 1991, J. Exp. Med. 173: 
1483-1491; Chappel et al., 1991, Proc. Natl. Acad. Sci. USA 88(20): 9036-9040; Chappel et 
al., 1993, Journal of Biological Chemistry 268:25124-25131; Tao et al., 1991, J . Exp. Med. 
173: 1025-1028; Tao et al., 1993, J. Exp. Med. 178: 661-667; Redpath et al., 1998, Human 
Immunology, 59, 720-727. Using methods known in the art, it is possible to determine 
corresponding or equivalent residues in proteins that have significant sequence or structural 
homology with each other. By the same token, it is possible to use such methods to 



58 



WO 2005/056759 PCT/US2004/040694 

engineer amino acid modifications in an antibody or Fc fusion that comprise constant regions 
from other immunoglobulin classes, for example as described in USSN 60/621,387 and 
60/629,068, to provide optimal properties. As an example, the relatively poor effector 
function of lgG2 may be improved by replacing key FcyR binding residues with the 
corresponding amino acids in an IgG with better effector function, for example lgG1 . For 
example, key residue differences between lgG2 and IgGI with respect to FcyR binding may 
include P233, V234, A235, -236 (referring to a deletion in lgG2 relative to lgG1), and G327. 
Thus one or more amino acid modifications in the parent lgG2 wherein one or more of these 
residues is replaced with the corresponding lgG1 amino acids, P233E, V234L, A235L, - 
236G (referring to an insertion of a glycine at position 236), and G327A, may provide 
enhanced effector function. Furthermore, one or more additional amino acid modifications, 
for example the S239D, V264I, A330L, I332E, or combinations thereof as described above, 
may provide enhanced FcyR binding and effector function relative to the parent lgG2. 
Figures 25a, 25d, and 25e illustrate this embodiment, providing the light and heavy chain 
sequences of AC1 0 variants that comprise L3.71/H3.70 AC10 combined with a number of 
possible variant lgG2 constant regions. 

[190] The Fc modifications defined in Figure 25 that provide enhanced effector function are 
not meant to constrain the invention to only these modifications for effector function 
optimization. For example, as described in US 6,737,056, PCT US2004/000643, USSN 
10/370,749, and PCT/US2004/005112, the substitutions S298A, S298D, K326E, K326D, 
E333A, K334A, and P396L provide optimized FcyR binding and/or enhanced ADCC. 
Furthermore, as disclosed in Idusogie et al., 2001, J. Immunology 166:2571-2572, 
substitutions K326W, K326Y, and E333S provide enhanced binding to the complement 
protein C1q and enhanced CDC. As described in Hinton et al., 2004, J. Biol. Chem. 279(8): 
6213-6216, substitutions T250Q, T250E, M428L, and M428F provide enhanced binding to 
FcRn and improved pharmacokinetics. Modifications need not be restricted to the Fc region. 
It is also possible that the mutational differences in the Fab and hinge regions may provide 
optimized FcyR and/or C1q binding and/or effector function. For example, as disclosed in 
USSN 60/614,944, and USSN 60/619,409, filed October 14, 2004, entitled "Immunoglobulin 
Variants Outside the Fc Region with Optimized Effector Function", the Fab and hinge 
regions of an antibody may impact effector functions such as antibody dependent cell- 
mediated cytotoxicity (ADCC), antibody dependent cell-mediated phagocytosis (ADCP), and 
complement dependent cytotoxicity (CDC). Thus immunoglobulin variants comprising 
substitutions in the Fc, Fab, and/or hinge regions are contemplated. For example, the 
antibodies may be combined with one or more substitutions in the VL, CL, VH, CH1, and/or 



59 



WO 2005/056759 



PCT/US2004/040694 



{ 



hinge regions. Furthermore, further modifications may be made in non lgG1 
immunoglobulins to corresponding amino acids in other immunoglobulin classes to provide 
more optimal properties, as described in USSN 60/621,387, filed 10/21/2004, entitled "IgG 
Immunoglobulin Variants with Optimized Effector Function". For example, in one 
embodiment, an lgG2 antibody, similar to the antibody presented in Figure 25, may comprise 
one or more modifications to corresponding amino acids in lgG1 or lgG3 CH1, hinge, CH2, 
and/or CH3. In another embodiment, an lgG2 antibody, similar to the antibody presented in 
Figure 25, may comprise all of the lgG1 CH1 and hinge substitutions,, i.e. said lgG2 variant 
comprises the entire CH1 domain and hinge of lgG1. 

Example 2. Immunogenicitv Reduction of C225 

[191] To illustrate further application of the method described in the present invention, and 
to validate its broad applicability to immunogenicity reduction of proteins, a second 
xenogeneic antibody example is provided using the variable region of C225 as the parent 
sequence (cetuximab, Erbitux®, Imclone) (US 4,943,533; PCT WO 96/40210). C225 is a 
murine anti-EGFR antibody, a chimeric version of which is currently approved for the 
treatment of cancer. A structural model of the murine C225 variable region was constructed 
using standard antibody modeling methods known in the art. Figures 26 and 27 show the 
sequences, host string content, and structures of the C225 VL and VH domains. A GDR 
graft of this antibody was constructed by placing the C225 CDRs into the context of the 
frameworks of the most homologous human germlines, determined to be vlk_6D-21 for VL 
and vh_4-30-4 for VH using the sequence alignment program BLAST. The sequences and 
string content of these CDR grafts are shown in Figures 28 and 29, along with structures of 
modeled C225 highlighting the mutational differences between the CDR grafted C225 
variable chains and WT. 

[192] Variants with reduced immunogenicity were generated by applying a string 
optimization algorithm on the WT C225 VL and VH sequences, similar to as described above 
for AC 10 except that single instead of multiple amino acid substitutions were sampled. HSC 
of each sequence was optimized using a window size w=9, and the same set of CDR and 
VLA/H interface proximal residues were masked. The calculation was run for C225 VL and 
VH in 1 00 separate iterations, generating a set of diverse C225 variants with more host 
string content than WT. Figure 30 shows the nonredundant set of output sequences from 
these calculations for the C225 VL and VH regions, referred to as C225 VL HSC Calculation 
1 and C225 VH HSC Calculation 1, respectively. In addition to the HSC score, the structural 
consensus and structural precedence of each sequence was evaluated (USSN 60/528,229, 



60 



WO 2005/056759 



PCT/US2004/040694 



filed December 8, 2003, entitled Protein Engineering with Analogous Contact Environments) 
in order to evaluate its structural integrity. 

[193] A second set of similar calculations were run on the C225 VL and VH sequences, 
except that the algorithm was allowed to sample multiple amino acid substitutions, rather 
than only single substitutions, in order to optimize HSC. Figure 31 shows the nonredundant 
set of output sequences from these calculations for the C225 VL and VH regions, referred to 
as C225 VL HSC Calculation 2 and C225 VH HSC Calculation 2, respectively. Here, two 
measure of structural fitness, referred to as "Structural Consensus" and "Structural 
Precedence" (USSN 60/528,229 and USSN 60/602,566), are used to evaluate the structural 
and functional integrity of the sequences, in addition to HSC score. The output sequences 
were clustered based on their mutational distance from the other sequences in the set, and 
these clusters are delineated by the horizontal black lines in the Figure. 

[194] The calculations described above and presented in Figures 30 and 31 were used to 
generate a set of C225 VL and VH variants. In some cases, further substitutions were made 
to output sequences, using string and structural scores, as well as visual inspection of the 
modeled C225 structure, to evaluate fitness. Figures 32 - 40 present the sequences, 
structural scores, string scores, and mapped mutational differences on the modeled C225 
structure for each of the C225 VL and VH variants. Iteration 21 from C225 VL HSC 
calculation 1 served as the precursor for L2 C225 VL, iteration 17 from C225 VL HSC 
calculation 2 served as the precursor for L3 C225 VL, and iteration 38 from C225 VL HSC 
calculation 2 served as the precursor for L4 C225 VL. Iteration 23 from C225 VH HSC 
calculation 1 served as the precursor for H3, H4, and H5 C225 VH, iteration 5 from C225 VH 
HSC calculation 2 served as the precursor for H6 C225 VH, iteration 41 from C225 VH HSC 
calculation 2 served as the precursor for H7 C225 VH, and iteration 44 from C225 VH HSC 
calculation 2 served as the precursor for H8 C225 VH. 

[195] Tables 7 and 8 present the mutational, structural fitness, and host string content 
scores for the C225 VL and VH variants as compared to the WT and CDR grafted C225 
sequences. In addition, the maximum identity match to the germline for each string in the 
sequences was also determined, referred to as N !D max. This represents the total number of 
strings in each sequence whose maximum identity to the corresponding strings in the human 
germline is the indicated value. For w = 9, Tables 7 and 8 list N 9 max, N 8 max, N 7 max, and 
N^max for each sequence. Also provided is the framework region homogeneity. In addition 
to the aforementioned structural and host string analysis, each sequence was analyzed for 
its global homology to the human germline; tables 7 and 8 present the most homologous 



61 



WO 2005/056759 



PCT/US2004/040694 



human germline sequence for each sequence (Closest Germline) and corresponding identity 
to that germline (ID to Closest Germline), determined using the sequence alignment program 
BLAST. 

Table 7. C225 VL Variants 





WT 


CDR 
Graft 


L2 


L3 


L4 


Mutations 




25 


17 


21 


18 


Structural 
Consensus 


0.49 


0.52 


0.56 


0.58 


0.54 


Oil UUIUI Cll 


0.53 


0.56 


0.57 






Precedence 


0.59 


0.57 


ri uiTiai 1 o in rig 

Content 


0.79 


0.94 


0.91 


0.92 


0.91 


Human String 
Similarity 


0.15 


0.65 


0.51 


0.58 


0.57 


Framework Region 
Homogeneity 




0.97 


0.52 


0.50 


0.78 


N 9 max 


13 


69 


52 


60 


58 


N 8 max 


27 


15 


28 


24 


23 


N 7 max 


37 


22 


20 


16 


19 


N«&max 


30 


1 


7 


7 


7 


Closest 
Germline 


6D-21 


6D-21 


3-11 


1D-13 


6D-21 


ID to Closest 


63/95 


87/95 


72/95 


73/94 


79/95 


Germline 


66% 


91% 


75% 


77% 


83% 



Table 8. C225 VH Variants 





WT 


CDR 
Graft 


H3 


H4 


H5 


H6 


H7 


H8 


Mutations 




33 


18 


21 


15 


21 


22 


28 


Structural 


0.44 


0.48 


0.51 












Consensus 


0.46 


0.49 


0.52 


0.49 


0.53 


Structural 
Precedence 


0.55 


0.54 


0.55 


0.54 


0.51 


0.55 


0.58 


0.55 


Human String 
Content 


0.67 


0.84 


0.79 


0.81 


0.77 


0.79 


0.79 


0.79 


Human String 
Similarity 


0.04 


0.56 


0.36 


0.41 


0.33 


0.36 


0.35 


0.33 


Framework Region 
Homogeneity 




0.97 


0.45 


0.52 


0.50 


0.50 


0.76 


0.77 


N 9 max 


3 


66 


42 


48 


38 


42 


41 


39 


N 8 max 


23 


17 


25 


24 


17 


24 


21 


27 


N 7 max 


32 


10 


19 


16 


26 


23 


25 


23 



62 



WO 2005/056759 



PCT/US2004/040694 



H8 

30 
3-33 

ID to Closest 56 / 99 88 / 99 67 / 99 74 / 99 64 / 99 69 / 99 67 / 99 80 / 99 
Germline 56% 88% 66% 74% 64% 70% 67% 81% 

[196] Again, whereas the CDR grafted C225 antibodies are most homologous to a single 
human germline sequence, the C225 variants of the present invention are homologous to 
different human germline sequences in different regions of the sequence. Whereas CDR 
grafted C225 VH is most homologous to human germline subfamily 4 across its entire 
sequence, H4 C225 VH is most homologous to subfamily 4 in FR1 , subfamily 3 in FR2, and 
subfamily 2 in FR3. Additionally, whereas the CDR grafted antibodies are most homologous 
to a single germline sequence that is also the most homologous sequence to the parent 
sequence, the present invention presents a set of antibodies for a given antibody that are 
most homologous to different human germline sequences, which need not be the most 
homologous germline sequence to WT. For example, Table 7 shows that CDR grafted C225 
VL is most homologous to vIk_6D-21 , which is also the most homologous human germline to 
WT C225. However L2, L3, and L4 are most homologous to three different human 
germlines - vlk>1 1, vlk_1D-13, and vlk_6D~21 respectively. Thus the variants of the 
present invention explore a substantially greater amount of diversity than CDR grafted 
antibodies. 

[197] The genes for the C225 variable regions were constructed as described above, and 
subcloned into a modified pASK84 vector (Skerra, 1994, Gene 141: 79-84) comprising 
mouse constant regions for expression as Fabs. Select C225 variants were experimentally 
tested for their capacity to bind EGFR antigen. L2/H3 and L2/H4 C225 Fabs were 
expressed from the pASK84 vector in E. Co// with a His-tag, and purified using Nickel-affinity 
chromatography. Antigen affinity of the C225 variants was tested using SPR similar to as 
described above. EGFR extracellular domain (purchased commercially from R&D Systems) 
was covalently coupled to the dextrane matrix of a CM5 chip using NHS-linkage chemistry. 
C225 Fabs were reacted with the EGFR sensor chip surface at varying concentrations. 
Global Langmuir fits were been carried out for the concentrations series using the 
BiaEvaluation curve fitting software. The on-rate constant (ka), off-rate constant (kd), 
equilibrium binding constant (KD=kd/ka), and predicted saturation binding signal (Rmax) 
derived from these fits are presented in Table 9, along with the Chi2 which quantifies the 



WT CDR H3 
Wl Graft 

N^max 61 26 33 

2EZ. «" "- 30 - 4 4 - M - 



H4 


H5 


H6 


H7 


31 


38 


30 


32 


2-26 


4-30-4 


3-33 


4-30-4 



63 



WO 2005/056759 



PCTAJS2004/040694 



average deviation of the fit curve from the actual data curve. The data indicate that both the 
L2/H3 and L2/H4 C225 variants bind EGFR antigen. 

Table 9. SPR data on C225 Variants 



C225 


ka (1/Ms) 


kd (1/s) 


KD(M) 


Rmax 
(RU) 


Chi2 


L2/H3 


2.79 x10 4 


5.35 X10 -3 


1.92x1 0" 7 


174 


8.83 


L2/H4 


1.79 x10 4 


4.73 X10- 3 


2.64 x It)" 7 


153 


2.69 



[198] In order to investigate the anti-EGFR variants in the context of a full length antibody, 
the C225 WT (LO ad HO) and variant (L2, L3, L4, H3, H4, H5, H6 ? H7, and H8) regions were 
subcloned into the mammalian expression vector pcDNA3.1Zeo (Invitrogen) as described 
above. All combinations of the light and heavy chain plasmids Were co-transfected into 293T 
cells, and antibodies were expressed, harvested, and purified as described above. Binding 
of the C225 WT (L0/HO) and variant (L0/H3, L0/H4, L0/H5, L0/H6, L0/H7, L0/H8, L2/H3, 
L2/H4, L2/H5, L2/H6, L2/H7, L2/H8, L3/H3, L3/H4, L3/H5, L3/H6, L3/H7, L3/H8, L4/H3, 
L4/H4, L4/H5, L4/H6, L4/H7, and L4/H8) antibodies was determined using SPR similar to as 
described above. Full length antibodies were flowed over the EGFR sensor chip described 
above. Figure 41 shows the SPR sensorgrams obtained from the experiments. The curves 
consist of a association phase and dissociation phase, the separation being marked by a 
little spike on each curve. As a very rough approximation the signal level reached near the 
end of the association phase can be used as an indicator for relative binding. For all the 
curves this signal level is within 25% of the average level indicating that none of the antibody 
variants have significantly lost their ability to bind to EGFR. 

[199] To assess the capacity of the anti-EGFR antibodies to mediate effector function 
against EGFR expressing cells, the C225 variants were tested in a cell-based ADCC assay. 
Human peripheral blood monocytes (PBMCs) were used as effector cells, A431 epidermoid 
carcinoma cells were used as target cells, and lysis was monitored by measuring LDH 
activity using the Cytotoxicity Detection Kit as described above. Figure 42 shows the dose 
dependence of ADCG at various antibody concentrations for WT and variant C225 
antibodies. The results show that a number of the C225 variants have comparable or better 
ADCC than WT C225 with respect to potency and efficacy. These data may be weighed 
together with the antigen affinity data and other data to choose the optimal anti-EGFR 
clinical candidate. As exemplified above with AC10 variants, combinations of the C225 
variants of the present invention with amino acid modifications that alter effector function are 



64 



WO 2005/056759 



PCTAJS2004/040694 



contemplated. 

Example 3. Immunogenicity Reduction of ICR62 

[200] To further illustrate application of the method described in the present invention, and 
to validate its broad applicability to immunogenicity reduction of proteins, an example is 
provided using as the parent sequence the anti-EGFR antibody ICR62 (Institute of Cancer 
Research) (PCT WO 95/20045; Modjtahedi et al., 1993, J. Cell Biophys. 1993, 22(1-3):129- 
46; Modjtahedi et al., 1993, Br J Cancer. 1993, 67(2):247-53; Modjtahedi et al, 1996, Br J 
Cancer, 73(2):228-35; Modjtahedi et al, 2003, Int J Cancer, 105(2):273-80). A structural 
model of the rat ICR62 variable region was constructed using standard antibody modeling 
methods known in the art. Figures 43 and 44 show the sequences, host string content, and 
structures of the ICR62 VL and VH domains. A CDR graft of this antibody was constructed 
by placing the ICR62 CDRs into the context of the frameworks of the most homologous 
human germlines, determined to be vlk_1-17 for VL and vh_1-f for VH using the sequence 
alignment program BLAST. The sequences and string content of these CDR grafts are 
shown in Figures 45 and 46, along with structures of modeled ICR62 highlighting the 
mutational differences between the CDR grafted ICR62 variable chains and WT. 

[201] Variants with reduced immunogenicity were generated by applying a string 
optimization algorithm on the WT ICR62 VL and VH sequences, similar to as described 
above for AC10 except that single instead of multiple amino acid substitutions were 
sampled. HSC of each sequence was optimized using a window size w=9, and the same 
set of CDR and VLA/H interface proximal residues were masked. The calculation was run 
for ICR62 VL and VH in 100 separate interactions, generating a set of diverse ICR62 
variants with more host string content than WT. Figure 47 shows the nonredundant set of 
output sequences from these calculations for the ICR62 VL and VH regions, referred to as 
ICR62 VL HSC Calculation 1 and ICR62 VH HSC Calculation 1 respectively. In addition to 
the HSC score, the structural consensus and structural precedence of each sequence was 
evaluated (USSN 60/528,229, filed December 8, 2003, entitled Protein Engineering with 
Analogous Contact Environments) in order to evaluate its structural integrity. 

[202] The calculations described above and presented in Figure 47 were used to generate 
a set of ICR62 VL and VH variants. In some cases, further substitutions were made to 
output sequences, using HSC and Structural Precedence scores, as well as visual 
inspection of the modeled ICR62 structure, to evaluate fitness. Figures 48 - 50 present the 
sequences, host string content, and mapped mutational differences on the modeled ICR62 
structure for each of the ICR62 VL and VH variants. Iteration 20 from ICR62 VL HSC 



65 



WO 2005/056759 



PCT/US2004/040694 



calculation 1 served as the precursor for L2 ICR62 VL Iteration 1 from ICR62 VH HSC 
calculation 1 served as the precursor for H9, and iteration 5 from ICR62 VH HSC calculation 
2 served as the precursor for H10 ICR62 VH. 

[203] Tables 10 and 11 present the mutational, structural fitness, and host string content 
scores for the ICR62 VL and VH variants as compared to the WT and CDR grafted ICR62 
sequences. In addition, the maximum identity match to the germline for each string in the 
sequences, referred to as N (D max, is also provided, as well as the framework region 
homogeneity. In addition to the aforementioned structural and host string analysis, each 
sequence was analyzed for its global homology to the host germline; tables 10 and 1 1 
present the most homologous host germline sequence for each sequence (Closest 
Germline) and corresponding identity to that germline (ID to Closest Germline), determined 
using the sequence alignment program BLAST, 

Table 10. ICR62 VL Variants 





WT 


CDR 
Graft 


L2 


Mutations 


0 


11 


6 


Structural 
Consensus 


0.56 


0.60 


0.61 


Structural 
Precedence 


0.52 


0.58 


0.57 


Human String 
Content 


0.86 


0.91 


0.90 


Human String 
Similarity 


0.38 


0.58 


0.56 


Framework Region 
Homogeneity 


0.62 


0.97 


0.64 


N 9 max 


37 


59 


56 


N B max 


26 


21 


19 


N 7 max 


31 


18 


22 


N^max 


13 


9 


10 


Closest 
Germline 


1-17 


1-17 


1-17 


ID to Closest 


76/95 86/95 81 /9( 


Germline 


80% 


90% 


85% 



Table 11. ICR62 VH Variants 

WT CDR Graft H9 H10 
Mutations 0 34 20 21 



66 



WO 2005/056759 



PCT/US2004/040694 





WT 


CDR Graft 


H9 


H10 


otructurai 
Consensus 


0.43 


0.44 


0.46 


0.45 


Structural 
rreceuence 


0.42 


0.52 


0.47 


0.49 


Human String 
oonient 


0.64 


0.85 


0.79 


0.79 


Human String 
Similarity 


0.01 


0.54 


0.28 


0.33 


Framework Region 
Homogeneity 




1.00 


0.64 


0.85 


N 9 max 


1 


64 


33 


39 


N 8 max 


16 


24 


33 


30 


N 7 max 


35 


14 


28 


25 


N^max 


67 


17 


25 


25 


Closest 
Germline 


1-f 


1-f 


1-f 


1-f 


ID to Closest 


60/98 


92/98 


72/98 


77/98 


Germline 


61% 


93% 


73% 


79% 



[204] Again, as observed from the significant differences in FRH and closest germlines, the 
ICR62 variants are homologous to different host germline sequences in different regions of 
the sequence. The genes for the ICR62 WT and L2/H9 variable regions were constructed 
as described above, and subcloned into a modified pASK84 vector (Skerra, 1994, Gene 141: 
79-84). The ICR62 Fabs experimentally tested for their capacity to bind EGFR antigen. WT 
and L2/H9 ICR62 Fabs were expressed from the pASK84 vector in E. Coli with a His-tag, 
and purified using Nickel-affinity chromatography. Antigen affinity of the ICR62 antibodies 
was tested using SPR similar to as described above, with EGFR covalently coupled to the 
CMS chip reacted with IGR62 antibodies at varying concentrations. The fits to the data, as 
described above, are provided in Table 12. fits were been carried out for the concentrations 
series using the BiaEvaluation curve fitting software. As can be seen, L2/H9 ICR62 binds 
with comparable affinity as WT to the EGFR antigen. 



Table 12. SPR data on ICR62 Variants 



ICR62 


ka 
(1/Ms) 


kd (1/s) 


KD (M) 


Rmax 
(RU) 


Chi2 


WT 


9.86 x 
10 4 


2.53x10" 

5 


2.57 x 10" 

10 


402 


1.86 


L2/H9 


2.35 x 
10 s 


1.06x10- 

4 


4.50 x 10- 

10 


508 


4.91 



67 



WO 2005/056759 

Example 4. String diversity exploration of immunoglobulins 



PCT/US2004/040694 



[205] The generation of mutational diversity based on HSC is much broader than the 
primary variant - secondary variant strategy described above for H3/L3 AC 10. Indeed 
substitutions can be designed for any parent protein wherein the substitutions result in 
positive or neutral impact on the host string content of the parent sequence. Again, the 
advantage of such a strategy is that it generates a diverse set of minimally immunogenic 
variants that have the potential for optimized properties, including but not limited to antigen 
affinity, activity, specificity, solubility, expression level, and effector function. Such a set of 
variants may be designed, for example, to explore diversity for other parent 
immunoglobulins, including but not limited to nonhuman antibodies, humanized or otherwise 
engineered antibodies (Clark, 2000, Immunol Today 21 :397-402), (Tsurushita & Vasquez, 
2004, Humanization of Monoclonal Antibodies, Molecular Biology of B Cells, 533-545, 
Elsevier Science (USA)), and "fully human" antibodies, obtained for example using 
transgenic mice (Bruggemann ef a/., 1 997, Curr Opin Biotechnol 8:455-458) or human 
antibody libraries coupled with selection methods (Griffiths ef a/., 1998, Curr Opin Biotechnol 
9:102-108). 

Example 5. Unique properties of variant proteins generated by the methods of the present 
invention 

[206] The methods described in the present invention generate variant proteins that 
possess a number of unique properties relative to variant proteins generated by other 
methods that attempt to achieve the same or similar goal. Figures 51 and 52 provide the 
host string content (HSC, Equation 3), exact string content (ESC, Equation 3a), and 
framework region homogeneity (FRH, Equation 10) of the AC10, C225, and ICR62 VH 
(Figure 51 ) and VL (Figure 52) variants of the present invention, compared with a number of 
antibody variable regions "humanized" by methods in the prior art. If a variant sequence's 
exact string content is derived solely from a single germline sequence, the FRH would be 
close to 1.0. Alternatively, as is the case with many of the variant sequences created by the 
present invention, FRH values can be significantly less than 1, with values ranging from 0.4 
to 1.0, indicating, as expected, that sequences with high exact string content can be 
discovered with contributions from multiple germline subfamilies and sequences. At the 
same time, the variant sequences engineered using the present invention have high host 
string content, and thus are predicted to have low potential for immunogenicity in humans. 
For example, as shown in Figure 51 variant VH sequences generated using the present 
invention have HSC values generally higher than 75%, and many of them have FRH values 



68 



WO 2005/056759 



PCT/US2004/040694 



lower than 0.6, indicating their HSC is derived from multiple germline frameworks. As shown 
in Figure 52, similar trends apply for variant VL sequences generated using the present 
invention 

[207] Whereas particular embodiments of the invention have been described above for 
purposes of illustration, it will be appreciated by those skilled in the art that numerous 
variations of the details may be made without departing from the invention as described in 
the appended claims. All references are herein expressly incorporated by reference. 



69 



WO 2005/056759 



PCT/US2004/040694 



CLAIMS 
We claim: 

1 . A method of generating a variant protein for a host as compared to a parent protein, 
comprising: 

A. comparing said parent protein sequence with two or more natural protein 
sequences from said host species; 

B. analyzing one or more amino acid strings of said parent protein sequence 
with a corresponding amino acid string of each natural protein sequence; and, 

C. substituting one or more amino acids of said parent protein sequence with a 
corresponding amino acid string of a natural protein sequence on an amino acid 
string by amino acid string basis. 

D. wherein said variant protein has increased host string content as compared to 
said parent protein, and, 

E. wherein said substituted amino acids include a first substitution in a first string 
from a first natural protein, and a second substitution in a second string from a 
second natural protein. 

2. The method according to claim 1, wherein said substituted amino acid is a 
consensus of two or more natural proteins. 

3. The method according to claim 1, wherein said host species is human. 

4. The method according to claim 1, wherein said parent protein family is murine. 

5. The method according claim 1, wherein said natural protein sequences are germline 
sequences. 

6. The method according to claim 1, wherein one of the substitutions is not the 
corresponding residue from the most homologous natural sequence. 

7. The method according to claim 5, wherein one of the substitutions is not the 
corresponding residue from a consensus of homologous natural sequences. 

8. The method according to claim 1, wherein one of the substitutions is made at a 
position that is not surface exposed. 

9. The method according to claim 1, wherein said variant protein is an antibody. 

10. The method according to claim 9, wherein one of the substitutions is not in a CDR. 



70 



• 



WO 2005/056759 



PCTAJS2004/040694 



1 1 . The method according to claim 9, wherein one of the substitutions is made at a 
position that is part of the VH/VL interface. 

12. The method according to claim 9, wherein said variant antibody comprises an affinity 
for an antigen no lower than about two-fold less than said parent protein. 

13. The method according to claim 9, wherein said variant antibody comprises an affinity 
for an antigen of about two-fold more than said parent protein. 

14. The method according to claim 9, whereiq said variant protein comprises a 
framework region identity as compared to a natural protein sequence of less than about 
85%. 

15. The method according to claim 9, wherein said variant protein comprises a 
framework region homogeneity of less than about 0.6. 

16. The method according to claim 9, wherein said variant antibody comprises no 
substitutions within about 5 Angstroms of a CDR residue. 

17. The method according to claim 9, wherein said antibody has modulated effector 
function as compared to said parent antibody. 

18. The method according to claim 17, wherein said effector function is ADCC. 

19. The method according to claim 1, wherein more than one variant protein is generated 
from said method to form a variant protein set. 

20. The method according to claim 19, wherein said variant protein set comprises at 
least two variant proteins that differ by more than about 5 amino acids. 

21 . The method according to claim 19, further synthesizing at least one variant protein. 

22. The method according to claim 1, wherein said analyzing one or more amino acid 
strings is not applied to a CDR region. 

23. The method according to claim 1, wherein said variant protein has at least one 
improved property is selected from a group consisting of solubility, stability, expression, 
affinity, immunogenicity, activity and functionality. 

24. The method according to claim 1, wherein said analyzing one or more amino acid 
strings includes theoretical binding to MHC agretopes. 

25. The method according to claim 24, wherein said string is nine amino acids in length. 

26. A computer readable memory to direct a computer to function in a specified manner 
comprising: 



71 



WO 2005/056759 



PCT/US2004/040694 



A. a comparison module to compare a parent protein sequence and two or more 
host natural protein sequences; 

B. an analyzing module to provide a comparison between a parent protein 
sequence and one or more natural protein sequences; 

C. a substitution module to replace one or more amino acids said parent protein 
sequence with a corresponding amino acid string of a natural protein sequence on an 
amino acid by amino acid basis, 

D. wherein said variant protein has increased host string content as compared to 
said parent protein. 

27. A method of generating a variant protein for a host as compared to a parent protein, 
comprising: 

A. comparing said parent protein sequence with two or more natural protein 
sequences; 

B. substituting amino acids in said parent protein sequence with amino acids in 
one or more natural protein sequences, wherein a substitution increases the 
sequence identity of said variant protein to a natural protein sequence as compared 
to said parent protein within a string, 

C. wherein said variant protein has increased host string content as compared to 
said parent protein. 

28. A method of generating a variant protein for a host as compared to a parent protein, 
comprising: 

A. comparing said parent protein sequence with two or more natural protein 
sequences from said host species; 

B. analyzing one or more amino acid strings of said parent protein sequence 
with a corresponding amino acid string of each natural protein sequence; and, 

C. substituting one or more amino acids of said parent protein sequence with a 
corresponding amino acid string of a natural protein sequence on an amino acid 
string by amino acid string basis, 

D. wherein said variant protein has increased host string content. 

29. The method according to claim 28, wherein said substituted amino acid is a 
consensus of two or more natural proteins. 



72 



WO 2005/056759 



PCTAJS2004/040694 



30. The method according to claim 28, wherein said host species is human. 

31. The method according to claim 28, wherein said parent protein is murine. 

32. The method according to claim 28, wherein said natural protein sequences are 
germline sequences. 

33. The method according to claim 28, wherein one of the substitutions is not the 
corresponding residue from the most homologous natural sequence. 

34. The method according to claim 33, wherein one of the substitutions is not the 
corresponding residue from a consensus of homologous natural sequences. 

35. The method according to claim 28, wherein one of the substitutions is made at a 
position that is not surface exposed. 

36. The method according to claim 28, wherein said variant protein is an antibody. 

37. The method according to claim 36, wherein one of the substitutions is not in a CDR. 

38. The method according to claim 36, wherein one of the substitutions is made at a 
position that is part of the VH/VL interface. 

39. The method according to claim 36, wherein said variant antibody comprises an 
affinity for an antigen no lower than about two-fold less than said parent protein. 

40. The method according to claim 36, wherein said variant antibody comprises an 
affinity for an antigen of about two-fold more than said parent protein. 

41. The method according to claim 36, wherein said variant protein comprises a 
framework region identity as compared to a natural protein sequence of less than about 
85%. 

42. The method according to claim 36, wherein said variant protein comprises a 
framework region homogeneity of less than about 0.6. 

43. The method according to claim 36, wherein said variant antibody comprises less than 
about 28 amino acid substitutions, as compared to a parent protein. 

44. The method according to claim 36, wherein said variant antibody comprises no 
substitutions within about 5 Angstroms of a CDR residue. 

45. . The method according to claim 36, wherein said antibody has modulated effector 
function as compared to said parent antibody. 

46. The method according to claim 45, wherein said effector function is ADCC. 



73 



WO 2005/056759 PCT7US2004/040694 

47. The method according to claim 28, wherein more than one variant protein is 
generated from said method to form a variant protein set 

48. The method according to claim 47, wherein said variant protein set comprises at 
least two variant proteins that differ by more than about 5 amino acids. 

49. The method according to claim 47, further synthesizing at least one variant protein. 

50. The method according to claim 28, wherein said strings are not applied to a CDR 
region. 

51. The method according to claim 28, wherein said variant protein has at least one 
improved property is selected from a group consisting of solubility, stability, expression, 
affinity, immunogenicity, activity and functionality. 

52. The method according to claim 28, wherein said analyzing one or more amino acid 
strings includes theoretical binding to MHC agretopes. 

53. The method according to claim 52, wherein said string is nine amino acids in length. 



74 



WO 2005/056759 



1/61 



PCT/US2004/040694 




0) 

o 
c 

Q> 

D" 
<D 
CO 



> 

<D 

c 



o 
O 
c 

W 

E 



3 

D) 



m 
u 

■3 



in 



D 

U 



P* 



H H H H H H 

CJ 8 6 D § § 
TO CO CO CO TO CO 

3 S 5 3 S 3 „ „ 

pjcOTOTOTOCOTOTO 
WTOTOPSTOTOTOTO 

TO TO TO TO Q TO. CO TO TO 
TOTOTOTOTOTOTOTOTOTO 

9||||||Sg| 



■3 



n 

p{ r> o 



PJ 
Cm 



i-i 

H O 




ooooooa 

" * CO CO W * 

J 

COTOTOTOTOCOriTOW 

ss§§s§§s§ 

COCOCOTOTOCOCOTOCO 



TO TO CO CO 

§ § § § 

CO TO CO TO 



p3 p3 Ctl 




^ £t! fx! 

p$ ps p: 



ma 

O t5 O 

TO CO TO 

>J J J 

o a a 




^ ^ J 4 ^ ^ 

fti * ft PC» ?c. 

a |a i s ^ 

o o o o o 
s £ 5 s 



w w w w 

J J ^ 





o o o 

a a a.. 

ooocooooo 



td w pq w w w 
j hi 3 j j p 

o o o o o 

a a a r " 

a a a on 

n» r* 
(u 
2= 



TO Q TO TO 

U* b* Ct* [i4 . . 

£ K 

"- '~ "l to 





Pi 



e CO CO TO CO' TO' TO TO QTOUCOCOOTCOTO 

ooooooooooooooouo 

"totgcgcgcgcgcgtgcgcg" *" " 

_ _ ; ; : _ : _ e 

cococococqtotototototocotototototo 
cococococqtotototototococototococo 



h3.v4 ^ »4 A k3 

CO TO TO CO CO _. - 

88888SS888 8888888888 

A ^ ^ a >i 

88888S 

o u o 5 o u 

TO CO TO TO CO CO 

assess 



5 W & W W M 



t>>>>>>>>>>M>>>> 

OOC30C?OOOC)> D O O O O U CD 

OCSOUUt&OOOOOOCOCDOOO 
COCOTOTOTOTOTOTOTOTOTOCOTOTOCOTOTO 

>>>>>>?s>£>>:>>>>> 
Hwwwooowwwwwtowtaiacfl 



eo ^» in u> eo cn loo 

oinoHN^xrujvo'H m in r- 

i i i i i i i t i i lit 

H | r, | r '| H | HHHHr,H N«, N , 



°l 01 1 m \ n | 00 1 1 



WO 2005/056759 PCT/US2004/040694 

2/61 



I 



g 

Q) 
T3 
U 

•a 



T3 
CD 
3 
C 



o 
o 



TO 



LL- 




PS 



8* Q 

CQ CO CO 

5 S 5 

ill 

So a _ _ _ 
cocotococococococo 



CO CO CO CO 
H H > H 
CO CO CO CO 

oooo 

CO CO CO CO 

o o u D 

ssss 

CO CO CO CO 

gee i3 





CO 






M 






3: 






S* 




CO 


CO 


CO 


FT 


FT 


vs 




CO 


CO 


55 




Q 




5 




CO 


CO 


CO 


KG 


KG 


M 


u 


u 


s 


CO 


CO 




5 


M 

s 


CO 








CO 


CO 


g 


w 


W 




o 




CO 




• i i i i i i i 



WO 2005/056759 



3/61 



PCT/US2004/040694 



to 
o 
o 
c 

CD 
3 

cr 
o 

CO 

__: 
-J 

> 

c 



o 
O 

c 

« 

E 

3 
X 



2 

3 

i? 
U. 



SH 
r- o 

r. crv 



s 

8 



Pi 

F. 



_ 

in 



Q) 

■a 

ro u 

■3 



r- 

VD 

in 




U 

— .03 

oia 



C. Ex* 



OICOOOCOCQCOCOCOCOCOCOCQCOCOCOCO 

M _______ _ 

H 
O 

W 
O 
CQ 
O 
CO 

& 

CO V_ v. w w_ _„ „, W_ w 

> o o o o 

iS 

) CO 

is 




& 

04 (. (_■ C-l C-| 

CO CO CQ CO CO 

CO CQ CO W CO 

M M M M H 

H H H H B 

r r r r r 

o o o o o 

CO CQ CO CO CO 

o o o o o 

CO CO CO PS CO 

O O _) O c_ 

CO CO CO CO CO 

l_, U* Cii fx. 

Pi pc! pi ps ps 

CO CO CO CO CO 

6 & > 2 _> 
o o o 

a. 

CO 
CO 



3 



__MHHMI.MMIH»HMh.MMI-H»-.l-.>-l 

t. b. S-i w 

OOOOOUO_>OC5C.OUOO_JC_> 

ssssasss&ssssssss 

r_ri<r.r_(_<__oaa_3toiaooaac. 



M M M i_| M 

. _ ^4 _ b 

_!____! 
si §! Si E. 04 
S S 6 B & 



CO 



o o 




BP 

CO CO 

o o 

CO CO 

P_ Cx. 

Pi Pi 

Q Q 



O CD O 

CO CO CO 

u o o 

CO CO CO 

o u o 

CO CO CO 



CO CO 

o o 

CO CO 

V) u 

CO CO 

ps 5 

Q P 

t> t> t> f> > 

CO CO cof 



Cx* P. Cti 

Pi Pi pi 

O Q Q 

_v g: 



o o o 

CO CO CO 

O CD o 

CO CO CO 

_)_)_) 
CO CO CO 



CO CO CO 

So > 

.M 

H H H 

H. H. 

O O O 

04 P. P* 

CO CO P. 

o o o» 
o o o 

ass 



-333-1 

oSSoo 

&l P» pLl &» Pi 

CO CO CO CO CO 
0 0 0 0 0 

_> O CD O _► 



u U U U U'U UUUUUUUUUUUUUUUU 
HHHCOHHHHHHHHHHHHHHHHMH 



O O 



8 



o a a a 



_ _ _ u i 

CO CO CO CO I 



8 



o a o 



q q a Q o 

I CO CO CO CO CO 

| [l* l-l t. > > 

I CO a &» CO 03 

I CO CO CO CO CO 

, Pi O* CU 0* Dj 

I CO CO CO CO CO 

t o o o o - 




M H h. M I 

QS50QQQGQQQ< 



QQQOQOQO 



1 i 



m i n vd r* to r- m (\ n 

eo .NH_H«>^l^H^n_^r.^nn.. 
Lnvocoic.Htr.tr.ir.tr.r.iroir.1 i i 

I I IQ> I Q I Q I Q l Q l > D i Q « Q Q Q 

si. . I. s* ^ -------- - - - 



II 



, co cn | o o 

-» c. oor.r.-.onO-' 

oj « r. i i r. n i kj> \ 

»Q » Q Q i » Q i P 

_'_' j j J J J j j J 



III I 



WO 2005/056759 



4/61 



PCT/US2004/040694 



/ 



a -9 

3 W> 



r- 

LD 



« r> o 



0) 



c 
o 

o 



3 



8 



CM 



8 



r-l 
OS 




h H H U) H O N 

O I H I H I CN I 

t a » Q i Q t Q 

n rt o o o f*) n ro 




WO 2005/056759 PCTYUS2 004/040694 

5/61 



Figure 1c 



9 10 

Rabat 678901234567 

IGKJ1 WTFGQGTKVEIR 

IGKJ2 YTFGQGTKLE I K 

IGKJ3 FTFGPGTKVDIR 

IGKJ4 LTFGGGTKVE IK 

IGKJ5 I TFGQGTRLE I K 

10 11 

Rabat 1234567890123 

IGHJ1 AEYFQHWGQGTLVTVSS 

IGHJ2 YWYFDLWGRGTLVTVSS 

IGHJ3 AFDVWGQGTMVTVSS 

IGHJ4 YFDYWGQGTLVTVSS 

IGHJ5 NWFDSWGQGTLVTVSS 

IGHJ6 YYYYYGMD WGQGTTVTVS S 



WO 2005/056759 PCT/US2004/040694 

6/61 



Figure 2a 



QVQLQQSGPE LVK PGASLKLSCT ASGFNI K 
QVQLVQS GAE VKK P GASVKVSCK ASGYT F T 

IDstringo=i5) = 6 



Figure 2b 

VH_m4D5 QVQLQQSGPELVK PGASLKLSCT ASGFNI K 

VH_l-2 QVQLVQS GAE VKKPGASVKVSCKASGYTFT 

VH_2-5 QITLKESGPTLVKETQTLTLTCTFSGFSLS 

VH_3~ 7 EVQLVE S GGGLVQ PGGSLRLSCAASGFT F S 

VH_4- 4 QVQLQESGPGLVKPPGTLSLTCAVSGGSI S 

VH_5- 51 EVQLVQS GAE VKKPGE SLKISCKGSGYS F T 

VH_6- 1 QVQLQQSGPGLVKPSQTLSLTCAI SGDSVS 

VH_7- 4- 1 QVQLVQSGSELKKPGASVKVSCKASGYTFT 



IDepltopeo=i5> = 6 
IDepltopeo-nD = 4 
IDepltopeo«i5) = 6 
IDepltopeo=i5) = 3 
IDepitbpecNis) = 6 
IDepitopeo»i5) = 3 
IDepltopeo=i5) = 6 

v v ^ 

IDmaxo=i5) = 6 



Figure 2c 



VH_m4D5 QVQLQQSGPE LVK PGASLKLSCTASGFN IK 

VH_l-2 QVQLVQS GAE VKK PGASVKVS CKASGYT F T 

VH_2- 5 QI TLK ESGPT LVK PTQTLTLTCTF SGF SLS 

VH_3- 7 EVQLVE S GGGLVQ PGG SLRLS CAASGFT F S 

VH_4- 4 QVQLQESGPGLVKPPGTLSLTCAVSGGSIS 

VH_5-51 EVQLVQSGAEVKKPGESLKI SCKGSGYSFT 

VH_6-1 QVQLQQS GPGLVK PS QTL SLT CAI SGDSVS 

VHJ7- 4- 1 QVQLVQS GSE LKK PGASVKV S CKASGYT F T 

IDo=i)max = 9 IDo=i2)max = 6 
IDo-2)max = 8 IDo=i3)max = 7 
IDo=3)max = 8 |Do-i4)max = 7 
IDo=^max = 8 lDo=i6)max = 6 
IDcr=5)max = 8 IDo=ie)max = 6 
lD(R8>max ~ 8 IDo=i7)max = 7 
. IDo=7ynax = 7 IDo=»i8)max = 7 
IE3(i=8)max - 7 IDo=i9)max = 7 
IDo=e)max = 7 IDo=20)max = 7 
ID(Rio)max = 7 IDo»2i)max = 6 
IDo=n)max = 7 lDo=*22)max = 5 



HSC(s) = 78.3 



WO 2005/056759 



7/61 



PCT/US2004/040694 



Figured L0AC10VL 



Figure 3a 

DIVLTQSPASLAVSLGQRATISCKASQSVDFDGDSYMNWYQQKPGQPPKVLIYAASNLESG 
IPARFSGSGSGTDFTLNIHPVEEEDAATYYCQQSNEDPWTFGGGTKLEIK 



Figure 3b 

FR1 FR2 FR3 FR4 




Figure 3c 

FR1 FR2 FR3 FR4 



una 

ux p. 

HTii ! 


T 
t. 


fiJ 


■ ASzr:.-..t-~. 

"i'it 

: i 


1 

■*■ 

T 




.CI 

...... 


:;uiixff 


tr j 

1H 


— 1 — mm 

* r Hff 

t . 


■""til ' !: 

p-lifTi 




:;Q 

4 1 -4 
* - p ■:- i 


i 1; >& 


; • •; I 


; 


1 








n 




il 


t •* nH 
I SH£ 


&.4.i!--.;|y-- 


iri" 


.:"4- l . j. 


i: ■■:]:: . >:.i 
: |= !•• 










it r ^"" 


rr-t 




i r * i 






: ! - • 






r.i_ -r 





















Figure 3d 



WO 2005/056759 



8/61 



PCT/US2004/040694 



Figure 4. H0AC10VH 



Figure 4a 

QIQLQQSGPEWKPGASVKISCKASGYTFTDYYITWVKQKPGQGLEWIGWIYPGSGNTKYN 
EKFKGKATLWDTSSSTAFMQLSSLTSEDTAVYFCANYGNYWFAYWGQGTQVTVSA 



Figure 4b 




Figure 4c 

FR1 FR2 FR3 FR4 







i !..L.L.g = 


eli. 


- ^ 


""1 

■ !-• 




-; j.! - . .;.;:_!■- 




l.r-L 


1 1 
t 






J 




. :.] 

• • -i 


[ j i j i 




in i ;!; 


If 




i 

j' 






1 : - 


$ 


• L 

i 


1 

i 




f 

T 




-;:.tT| 

i « I 

Itl 






-r t--i-!. %ri=Hi 

*~ + » 1 "* J ■ iT 








M 


j_ 
















■ ■. : i n rr i ; 




i I 




; > ; i 1 1 i t 






~T~! 



Figure 4d 



WO 2005/056759 



9/61 



PCT/US2004/040694 



Figures. CDR grafted AC10 VL 



Figure 5a 

DIVMTQSPDSLAVSLGERATINCKSSQSVDFDGDSYLAWYQQKPGQPPKLLIYAASNLESG 
VPDRFSGSGSGTDFTLTISSLQAEDVAVYYCQQSNEDPWTFGGGTKLEIK 



FR1 



Figure 5b 

FR2 



FR3 



FR4 



Figure 5c 

FR1 FR2 FR3 FR4 




Figure 5d 



WO 2005/056759 



10/61 



PCT/US2004/040694 



Figure 6. CDR grafted AC10VH 



Figure 6a 

QVQLVQSGAEVKKPGASVKVSCKASGYTFTDYYMHWVRQAPGQRLEWMGWIYPGSGNT 
KYSQKFQGR\n"ITRDTSASTAYMELSSLRSEDTAVYYCARYGNYWFAYWGQGTLVTVSS 



Figure 6b 




Figure 6d 



WO 2005/056759 



11/61 



PCT/US2004/040694 




WO 2005/056759 



12/61 



PCTAUS2004/040694 



Figure 8. L1 AC10VL 



Figure 8a 

DIVLTQSPATLSLSPGERATLSCRASQSVDFDGDSYMNWYQQKPGQPPKVLIYAASNLES 
GIPARFSGSGSGTDFTLTISSLQPEDFATYYCQQSNEDPWTFGGGTKVEIK 



FR1 



Figure 8b 

FR2 



FR3 




S IIM 



FR4 




FR1 



Figure 8c 

FR2 



FR3 



FR4 



















rT • ii'TiriM— i •! r~' 






r^ft ■ 




e 

p — [ 






:'C:|. 

! 

, "i 




Li. MJ ..J..:..; i. .. 

;-r|=n- itr -!-"-; 










ti- ! 






i i 


Mr- tip 








- aa--!~ 




M _ -: 










; K~ : -Vj- '{I 










_L..t ~ 





Figure 8d 




WO 2005/056759 



13/61 



PCTAJS2004/040694 



Figure 9. L2 AC10VL 



Figure 9a 

DIVLTQSPSSLSASVGDRVTITCRASQSVDFDGDSYMNWYQQKPGQPPKVLIYAASNLESG 
IPARFSGSGSGTDFTLTISSLQPEDFATYYCQQSNEDPWTFGGGTKVEIK 



Figure 9b 

FR1 FR2 FR3 FR4 




Figure 9d 



WO 2005/056759 



14/61 



PCT/US2004/040694 



Figure 10. L3AC10VL 



Figure 10a 

DIVLTQSPDSLAVSLGERATINCKASQSVDFDGDSYMNWYQQKPGQPPKVLIYAASNLESG 
IPARFSGSGSGTDFTLTINSLEAEDAATYYCQQSNEDPWTFGGGTKVEIK 



Figure 10b 

FR1 FR2 FR3 FR4 





Figure 10d 



WO 2005/056759 



15/61 



PCT/US2004/040694 



Figure 11. H1 AC10VH 
Figure 11a 

QIQLVQSGPEVKKPGASVKVSCKASGYTFTDYYITWVRQAPGQGLEWMGWIYPGSGNTK 
YNEKFQGRVTITVDTSASTAYMELSSLRSEDTAVYFCANYGNYWFAYWGQGTLVTVSS 



Figure 11b 




Figure 11c 

FR1 FR2 FR3 FR4 









1 1 
























...UBS 




sar 


\ 




~J ' 








-U 




„l.. 


— — 










ti 






hJ 


iiiR 




j.. j. 










f If 4 :-H 












. .. . 




-"•-{ 


f-ix^3|Z^:. j-rr-j-- -ft:" 




— ^ 

II 






s 






^■ 




— \ -r- 




* t :-\ v: ; . r 4, - i - "7. i- 


— t-i •* i 









Figure 11d 



WO 2005/056759 



16/61 



PCT/US2004/040694 



Figure 12. H2AC10VH 
Figure 12a 

QIQLVESGGGLVKPGGSLRLSCAASGYTFTDYYITWVRQAPGQGLEWMGWIYPGSGNTK 
YNEKFQGRVTMTVDTSTSTAYMELSSLRSEDTAWFCANYGNYWFAYWGQGTLV7VSS 



Figure 12b 

FR1 FR2 FR3 FR4 




Figure 12c 

FR1 FR2 FR3 FR4 





! i l! j i" 


1 1 • 




: 1 


I • ' 




- 














J '■ 




1 1. 




r-^-fi:: r -^fi- 










| 

T..TZZ 












:.- . 


f sr -sr. \ :>-*• 
? \ * 


*■ f 

" ""I 


T" 
















-i • i i-i-: -i 1-+ 


-H 




- i 













Figure 12d 



WO 2005/056759 



17/61 



PCT/US2004/040694 



Figure 13. H3AC10VH 



Figure 13a 

QIQLVQSGPEVKKPGASVKVSCKASGYTFTDYYITWVRQAPGQGLEWMGWIYPGSGNTK 
YNEKFQGRFVFSVDTSASTAYLQISSLKAEDTAVYFCANYGNYWFAYWGQGTLVTVSS 



FR1 



FR2 



Figure 13b 



FR3 



FR4 




FR1 



FR2 



Figure 13c 



FR3 



FR4 







i 


r 

• 4 


'i — 

j 
! 




r --. • r 
!:~ ! 




M 11 


1 i !; ! 






« 






j. 

f- 




S.S 

r; 


i 

... ..... 


J. . 

V. 


..!..... 


■m 






...!_. .:.:«..! 
i :■ i- ! 






— 1 — 1 


ill. i't:' ! . 




i 


til IS; 




; h 




1 


i : i 1 


! if! 










J . 


■ ii rr™'" t- 




i i * 


!■ . , 1 


H ' • . 






i 





Figure 13d 



WO 2005/056759 



18/61 



PCT/US2004/040694 



Figure 14a 



100H 

o 

u 

§ 80 

(/) 

0) 

c 



E 

3 
—I 

E 

s 

5 



60 



40 



20- 




1 1 1 1 1 1 1 r 1 — 

-11 -10 -9 -8 -7 
log [antibody] (0.2M> 



Figure 14b 



iooH 

o 
u 

§ 80 

CO 
<D 

c 



E 

3 



60 



E 40 
X 



20- 




-12 



— i — 

-11 



i 

-10 



-9 



— i— 

-8 



-7 



— T— 

-6 



log [antibody] (0.2M) 



WO 2005/056759 



19/61 



PCT/US2004/040694 




WO 2005/056759 



20/61 



PCT/US2004/040694 




WO 2005/056759 



22/61 



PCT/US2004/040694 



Figure 17a 




O 1 2 

log [antibody] (ng/ml> 



□ HOLD 
O H0L1 
O H0L2 
A H0L3 
s H1L0 
o H1L1 
o H1L2 
A H1L3 



Figure 17b 




0 1 2 

tog [antibody] (ng/ml) 



WO 2005/056759 



23/61 



PCT/US2004/040694 




WO 2005/056759 



24/61 



PCTAJS2004/040694 




WO 2005/056759 



25/61 



PCT/US2004/040694 



Figure 18b 

35-i 




log [antibody] (ng/ml) 



■ H0L0 

• H3L3 

Q H3L3, 1332E 

H3L3, V264I/I332E 

* H3L3, S239D 

A H3L3, S239D/I332E 
V H3L3 

S239D/A330UI332E 



WO 2005/056759 



26/61 



PCT/US2004/040694 



Figure 19 




log [antibody] (0.1M) 



i 



WO 2005/056759 



27/61 



PCT/US2004/040694 



Figure 20. L3.71 AC10VL 



Figure 20a 

EIVLTQSPDSLAVSLGERATINCKASQSVDFDGDSYLNWYQQKPGQPPKVLIYAASTLQSG 
VPSRFSGSGSGTDFTLTINSLEAEDAATYYCQQSNEDPWTFGGGTKVEIK 



Figure 20b 

FR1 FR2 FR3 FR4 




Figure 20c 

FR1 FR2 FR3 FR4 




Figure 20d 



WO 2005/056759 



28/61 



PCT/US2004/040694 



Figure 21. L3.72AC10VL 



Figure 21a 

AIVLTQSPDSLAVSLGERATINCKASQSVDFDGDSYLNWYQQKPGQPPKVLIYAASTLETG 
VPSRFSGS GSGTD FTLTI N SLEAE DAATYYCQQS NEDPWTFGGGTKVE IK 



Figure 21b 

FR1 FR2 FR3 FR4 




Figure 21 d 



WO 2005/056759 



PCT/US2004/040694 



29/61 



Figure 22. H3.68 AC10 VH 



Figure 22a 

QLQLVQSGPEVKKPGASVKVSCKVSGYTFTDYYITWVRQAPGQALEWMGWIYPGSGNTK 
YNEKFQGRFVFSVDTSASTAYLQISSLKAEDTAVYFCANYGNYWFAYWGQGTLV7VSS 



FR1 



FR2 



Figure 22b 



FR3 



FR4 




FR1 



FR2 



Figure 22c 



FR3 



FR4 




-T-T.- 



-rr-r 




fir 



r---: — H--H-T- 



TTT 



■ I « 



llTBP, j 

1I_L 



Figure 22d 



WO 2005/056759 PCT/US2004/040694 

30/61 



Figure 23. H3.69AC10VH 



Figure 23a 

QLQLVQSGAEVKKPGASVKVSCKVSGYTFTDYYITVVVRQAPGQALEWMGWIYPGSGNTK 
YSQKFQGRFVFSVDTSASTAYLQISSLKAEDTAVYYCANYGNYWFAYWGQGTLVTVSS 



Figure 23b 

FR1 FR2 FR3 FR4 




Figure 23c 

FR1 FR2 FR3 FR4 





._L_L__U: •. ! ! 


•4--:-- 
VT " 


'■ \ ' 

_1lJ_: 


! 


— i-^- - -I !!" 

T-; r ;]|! 


; — 1 

i i 
L 1 




m ■ 


! .. ! li.i 
















- .Hi 







i { » "" " ; >l?i t 1m - , > ™ i i I > ■ "i ■ r " f ' 7~ » i I • ; »i ■ 



Figure 23d 



WO 2005/056759 



31/61 



PCT7US2004/040694 



Figure 24. H3.70AC10VH 



Figure 24a 

QLQLVQSGAEVKKPGASVKVSCKVSGYTFTSYYISWVRQAPGQALEWMGWIYAGSGNTK 
YSQKFQGRFVFSVDTSASTAYLQISSLKAEDTAVYYCANYGNYWFAYWGQGTLVTVSS 



Figure 24b 

FR1 FR2 FR3 FR4 




Figure 24c 

FR1 FR2 FR3 FR4 





BBC 










yf---. : 






: 5 . ! r.: 








I . I L . . 


iHI 


) 


t * 


T Oi i 

L ~ . ! - - . J_ 


i 




■ J ; 


rr---4 
. ill ;. i 






-r- >- - 

-jr.- -;-.}•_■ 

|._Uu 






- * •! 


• J • |->rr;^.th_.:-: r\ 






•»-!•: •' ;- • |4'- 




• 1 " 


HI- p.- .:■ 














;» t 




1 ! 


" 1 . 




i i: , 




i 





Figure 24d 



WO 2005/056759 



32/61 



PCT/US2004/040694 



Figure 25. Optimized Anti-CD30 lgG1 Antibodies 



Figure 25a. Anti-CD30 Light Chain 

EIVLTQSPDSLAVSLGERATINCKASQSVDFDGDSYLNWYQQKPGQPPKVLIYAASTLQSG 
VPSRFSGSGSGTDFTLTINSLEAEDAATYYCQQSNEDPWTFGGGTKVEIK RTVAAPSVFIFP 
PSDEQLKSGTASWCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTL 
TLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC 

Figure 25b. Anti-CD30 IgG1 Heavy Chain Comprising Possible Fc Variants 

QLQLVQSGAEVKKPGASVKVSCKVSGYTFTSYYISWVRQAPGQALEWMGWIYAGSGNTK 

YSQKFQGRFVFSVDTSASTAYLQISSLKAEDTAVYYCANYGNYWFAYWGQGTLVTVSSA S 

TKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLY 

SLSSWTVPSSSLGTQTYICNVNHKPSNTKVDKKAEPKSCDKTHTCPPCPAPELLGGPX,VF 

LFPPKPKDTLMISRTPEVTCWXzDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYR 

WSVLTVLHQDWLNGKEYKCKVSNKALPX3PX4EKTISKAKGQPREPQVYTLPPSRDELTKN 

QVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGN 

VFSCSVMHEALHNHYTQKSLSLSPGK 



Position 


EU Index 
Position 


WT 


Possible Substitutions 




239 


S 


D, E, N, Q, T 


x 2 


264 


V 


I.T.Y 


x 3 


330 


A 


Y.L.I 


X4 


332 


I 


D, E, N, Q 



Figure 25c. Anti-CO30 Fc Variant lgG1 Heavy Chain 

QLQLVQSGAEVKKPGASVKVSCKVSGYTFTSYYISWVRQAPGQALEWMGWIYAGSGNTK 

YSQKFQGRFVFSVDTSASTAYLQISSLKAEDTAVYYCANYGNYWFAYWGQGTLVTVSSA S 

TKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLY 

SLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKAEPKSCDKTHTCPPCPAPELLGGPDVF 

LFPPKPKDTLMISRTPEVTCVWDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYR 

WSVLTVLHQDWLNGKEYKCKVSNKALPLPEEKTISKAKGQPREPQVYTLPPSRDELTKNQ 

VSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNV 

FSCSVM HEALHNHYTQKSLSLSPGK 



WO 2005/056759 



33/61 



PCT/US2004/040694 



Figure 25d. Anti-CD30 lgG2 Heavy Chain Comprising Possible Fc Variants 

QLQLVQSGAEVKKPGASVKVSCKVSGYTFTSYYISWVRQAP6QALEWMGWIYAGS6NTK 

YSQKFQGRFVFSVDTSASTAYLQISSLKAEDTAVYYCANYGNYWFAYWGQGTLVTVSSA S 

TKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVtVSWNSGALTSGVHTFPAVLQSSGLY 

SLSS\A/TVPSSNFGTQTYTCNVDHKPSNTKVDKTVERKCCVECPPCPAPZ 1 Z 2 Z3Z 4 GPX 1 VF 

LFPPKPKDTLMISRTPEVTCWXzDVSHEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTFR 

WSVLTVVHQDWLNGKEYKCKVSNKZ5LPX3PX4EKTISKTKGQPREPQVYTLPPSREEMTK 

NQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGSFFLYSKLTVDKSRWQQG 

NVFSCSVMHEALHNHYTQKSLSLSPGK 



Position 


EU Index 
Position 


WT 


Possible Substitutions 




239 


S 


D, E, N, Q, T 


x 2 


264 


V 


I, T, Y 


x 3 


330 


A 


Y, L,l 


x 4 


332 


I 


D, E, N, Q 


Z1 


233 


P 


E 


z 2 


234 


V 


L 


z 3 


235 


A 


L 


Z4 


236 




G 


z 5 


327 


G 


A 



Figure 25e. Anti-CD30 Fc Variant lgG2 Heavy Chain 

QLQLVQSGAEVKKPGASVKVSCKVSGYTFTSYYISWVRQAPGQALEWMGWIYAGSGNTK 

YSQKFQGRFVFSVDTSASTAYLQISSLKAEDTAVYYCANYGNYWFAYWGQGTLVTVSSA S 

TKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLY 

SLSSVVTVPSSNFGTQTYTCNVDHKPSNTKVDKTVERKCCVECPPCPAPELLGGPDVFLFP 

PKPKDTLMISRTPEVTCVWDVSHEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTFRWS 

VLTWHQDWLNGKEYKCKVSNKGLPLPEEKTISKTKGQPREPQVYTLPPSREEMTKNQVS 

LTCLVKGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGSFFLYSKLTVDKSRWQQGNVFS 

CSVMHEALHNHYTQKSLSLSPGK 



WO 2005/056759 



34/61 



PCT/US2004/040694 



Figure 26. C225WTVL 
Figure 26a 

DILLTQSPVILSVSPGERVSFSCRASQSIGTNIHWYQQRTNGSPRLLIKYASESISGIPSRFSG 
SGSGTDFTLSINSVESEDIADYYCQQNNNWPTTFGAGTKLELK 



Figure 26b 




Figure 26c 

FR1 FR2 FR3 FR4 

?.•!». .i .... . if..... 



























> ' T" ~ V ~ 


in:"": 


itr 


u 

y. 


1- 

1 


~*"T' ~ 


TLTi:- 

im. 

! i I r 


•LfLl 

-Li L 




liilllilHi ! 

■E' 

"■wrap i - • 


j_: 

r - 
i 




" i t 






?r 






±,.. r r , t j 


it i 




t.-ti !l 


; ! -4 




tH|pa~"r.' 


i . 

i- 


j I- • i 


- •; f 

s . 1 






i ' 


r..~ i 


h-- 
i 




■n---- 




.L 






i 






J_ 










■ — i " 


i.~ 
' " 








""f r 






- : • ■«-■-■■. — |>r . 


-..-i - . .1- 


! . i 





Figure 26d 



WO 2005/056759 



35/61 



PCT/US2004/040694 



Figure 27. WTC225VH 
Figure 27a 

QVQLKQSGPGLVQPSQSLSITCTVSGFSLTNYGVHWVRQSPGKGLEWLGVIWSGGNTDY 
NTrPFTSRLSINKDNSKSQVFFKMNSLQSNDTAIYYCARALTYYDYEFAYWGQ 

Figure 27b 




FR1 



FR2 



Figure 27c 



FR3 



FR4 







ii i » 


', ■"' I ' 




















|: « ! 

• ; > ■ j. . 


i ! 1 






i 




: : - | ; : ; s 


- i; i 

■ 1 ! i 






-r 






i . i 


ill.:.. 

in 






! i 


r 1 

1 




i Mi i 






• i-.; 

--.i.ri 
-t-1 
. i 




; | r 


!>: s : I 


i* • 




1 ! 




f 


' ' * It 








! 






1 1 « ' t 






i 


i t ! ■ . • , i ii • i : . 


• j- 






L -!—t 



Figure 27d 



WO 2005/056759 



36/61 



PCT/US2004/040694 



Figure 28. CDR grafted G225 VL 
Figure 28a 

EIVLTQSPDFQSVTPKEKVTITCRASQSIGTNLHWYQQKPDQSPKLLIKYASESISGVPSRF 
SGSGSGTDFTLTINSLEAEDAATYYCHQNNNWPTTFGAGTKLEIK 



FR1 



Figure 28b 

FR2 



FR3 



FR4 





Figure 28d 



WO 2005/056759 



37/61 



PCTAJS2004/040694 



Figure 29. CDR grafted C225 VH 
Figure 29a 

QVQLQESGPGLVKPSQTLSLTCTVSGFSLTNYGWSWIRQPPGKGLEWIGYIWSGGNTYYN 
PSLKSRVTISVDTSKNQFSLKLSSVTAADTAVYYCARALTYYDYEFAYWGQGTLVTVSS 



FR1 




Figure 29b 

FR2 FR3 



HS ||l|ip; 



FR4 




FR1 



Figure 29c 



FR2 



FR3 



FR4 



' i; ; -■ 



- • ; 



:.=.! II. 



XX 



u 4* 



I l : i 



TP 



Figure 29d 



WO 2005/056759 PCT/US 2 004/040694 

38/61 



c 
o 



3 
O 

Ctt 

O 

O 
UJ 
X 

> 

m 

CM 
CN 

o 

CO 

o 

CO 

£ 



s r 



8 ,tt a a ■ a'aav'a m a a!aa km a'a.* a a KVaja 



e «. 



■■a <•» »j* M a"a|a w aja aja.a a a «<j»r|w+» ■ i 
ttsgngo vt^v^tuxt** aa>» 



IT 



:Tfc:yi:|: 



5: 

a ► 



6. * 

t a 



n. 



EM 



tanitli, 

m'm. «y a sjs *Va a a vSrxk ■ 



o.oooooooooeooaooo 



o » e.0.0 to A ( a a a n o» oso» o aoooot 




lip] i 



5 fc. 



a o ( a o;o « o t> o o o o o o a » a o ft-'» »o o] 



8!:' 



» a-3 a a z a a a * a a a z* * a-a ci a 

mum 

TTTT*i'» I » V IT TYH I iTT 



m I",*,* a aja <n as a a 

" oom« u*w'wVt> • * » u'» »'» uSa>* «|a 't» 



r 5. 

! I 



■ * ^ • a a t» *%*.a a » b a- »a a aa a a a a r 



»o * • a o t> o o o • o o o • • • e.a c*o » i 
• a** n*a« • a eTav a'* a a a a'a a a ea'a •*• • c 



:53r5!555£5:2H555555SS.l.: 
j { - SjlKSH = 3H552. t . : 35..3322£ 



iS 

3 

Ja> 

CO 

O 

O 
UJ 
X 

X 

> 

m 

CM 
CM 

o 



CO 



3 
O) 



r. . 

■ a! 

i v 

f [• 



t»ec'tu«t 



to 



i i * » i . i ♦ , i 
i;:i!:tl;i 



-a '» >>°>»i 



<I3!S!!5sn 



WO 2005/056759 



39/61 



PCT/US2004/040694 




WO 2005/056759 



40/61 



PCT/US2004/040694 



Figure 32. L2 C225 VL 
Figure 32a 

DILLTQSPATLSLSPGERVTLSCRASQSIGTNIHWYQQKPDQSPKLLIKYASESISGIPSRFS 
GSGSGTDFTLTISSLQPEDFADYYCQQNNNWPTTFGAGTKLEIK 



Figure 32b 

FR1 FR2 FR3 FR4 




Figure 32d 



WO 2005/056759 



41/61 



PCT/US2004/040694 



Figure 33. L3C225VL 
Figure 33a 

DILLTQSPSSLSASVGDRVTITCRASQSIGTNIHWYQQKPDQSPKLLIKYASESISGIPSRFS 
GSGSGTDFTLTISSLQAEDVAVYYCQQNNNWPTTFGAGTKLEIK ' " 



Figure 33b 




WO 2005/056759 



42/61 



PCT/US2004/040694 



Figure 34. L4C225VL 
Figure 34a 

DILLTQSPAFLSVTPGEKVTITCRASQSIGTNIHWYQQKPDQSPKLLIKYASESISGIPSRFSG 
SGSGTDFTLTINSLEAEDAATYYCQQNNNWPTTFGAGTKLEIK 



Figure 34b 

FR1 FR2 FR3 FR4 




Figure 34d 



WO 2005/056759 



43/61 



PCT/US2004/040694 



Figure 35. H3C225VH 
Figure 35a 

QVQLQQSGPGLVKPSQTLSLTCTVSGFSLTNYGVHWVRQAPGKGLEWLGVIWSGGNTDY 
NTSLKSRLTISKDNSKSQWLQMNSLRAEDTAVYYCARALTYYDYEFAYWGQGTLV7VSS 



Figure 35b 




Figure 35d 



WO 2005/056759 



44/61 



PCT/US2004/040694 



Figure 36. H4C225VH 
Figure 36a 

QVQLQQSGPGLVKPSQTLSLTCTVSGFSLTNYGVHWVRQAPGKGLEWLGVIWSGGNTDY 
NTSLKSRLTISKDNSKSQWLTMTNMDPVDTATYYCARALTYYDYEFAYWGQGTLVTVSS 



Figure 36b 

FR1 FR2 FR3 FR4 




Figure 36c 

FR1 FR2 _ FR3 F* 4 




Figure 36d 



WO 2005/056759 



45/61 



PCT/US2004/040694 



Figure 37. H5C225VH 
Figure 37a 

QVQLQQSGPGLVKPSQTLSLTCTVSGFSLTNYGVHWVRQAPGKGLEWLGViWSGGNTDY 
NTPLTSRLTINKDNSKSQWLQMNSLRAEDT^ 



Figure 37b 

FR1 FR2 FR3 FR4 




Figure 37c 

FR1 FR2 FR3 FR4 




Figure 37d 



WO 2005/056759 



46/61 



PCT/US2004/040694 



Figure 38. H6C225VH 
Figure 38a 

QVQLQQSGPGLVKPSQTLSLTCTVSGFSLTNYGVHWVRQAPGKGLEWVGVIWSGGNTDY 
NTSVKGRFTISKDNSKSQWLQMNSLRAEDTANAYCARALTYYDYEFAYWGQGTLVTVSS 

Figure 38b 




FR4 



FR1 



FR2 



Figure 38c 



FR3 



FR4 




Figure 38d 



WO 2005/056759 



47/61 



PCT/US2004/040694 



Figure 39. H7C225VH 
Figure 39a 

QVQLQQSGPGLVKPSQTLSLTCTVSGFSLTNYGVHWVRQPPGKGLEWIGVIWSGGNTDY 
NTSLKSRVTISKDNSKSQVSLKLSSVTAADTAVYYCARALTYYDYEFAYWGQGTLVTVSS 



Figure 39b 




Figure 39d 



WO 2005/056759 



48/61 



PCT/US2004/040694 



Figure 40. H8C225VH 
Figure 40a 

QVQLVESGGGLVQPGRSLRLSCAVSGFSLTNYGVHWVRQAPGKGLEWVSVIWSGGNTD 

YNTSVKGRFTISKDNSKSTWLQMNSLRAEDTAVYYCARALTYYDYEFAYWGQGTLVW 

S 



FR1 



FR2 Figure 40b FR3 



FR4 




FR1 




Figure 40c 

FR2 FR3 



FR4 



Figure 40d 



WO 2005/056759 



49/61 



PCT/US2004/040694 




Time 



WO 2005/056759 



50/61 



PCT/US2004/040694 



Figure 42a 




log [antibody] (ug/ml) 




WO 2005/056759 



51/61 



PCT/US2004/040694 



100- 



80- 



o 
o 

< 60- 



w 40H 



20- 
0- 

^1.0 



Figure 42c 




i 

-3.5 



— l — 

-3,0 



— i — 

-2.5 



— , — 

-2.0 



-1.5 



— I — 

-1.0 



log [antibody] (ug/ml) 



Figure 42d 



— i — 

-0.5 



— I 

0.0 



■ H0L2 

♦ H0L3 

• H3L3 
a H4L3 
b H6L3 
+ H7L3 
© H8L3 



100- 



80- 

O 
O 
Q 
< 

CO 

E 

« 40H 



60- 



20- 
0- 

-4.0 




i 

-3.5 



—| — 

-3.0 -2.5 -2.0 -1.5 -1.0 
log [antibody] (ug/ml) 



—i — 

-2.0 



— i — 

-1.5 



-0.5 



— i 

0.0 



WO 2005/056759 



52/61 



PCT/US2004/040694 



Figure 43. WTICR62VL 
Figure 43a 

DIQMTQSPSFLSASVGDRVTINCKASQNINNYLNWYQQKLGEAPKRLIYNTNNLQTGIPSRF 
SGSGSGTDYTLTISSLQPEDFATYFCLQHNSFPTFGAGTKLELK 



Figure 43b 

FR2 




Figure 43c 











FR2 














SE 

'"■">' i "I ;" 


1 

i . 


i 










-h -fT 


1 : r 


BIT 


!-: :-;.*-h -I Mi 




_J= r : 1 ' 












i : 
1. i 





FR4 




Figure 43d 



WO 2005/056759 



53/61 



PCT/US2004/040694 



Figure 44. WTICR62VH 
Figure 44a 

QVNLLQSGAALVKPGASVKLSCKGSGFTFTDYKiHWVKQSHGKSLEWIGYFNPNSGYSTY 
NEKFKSKATLTADKSTDTAYMELTSLTSEDSATYYCTRLSPGGYWMDAWGQGASVTVSS 



Figure 44b 



FR1 



FR2 



FR3 



FR4 




FR1 



FR2 



Figure 44c 



FR3 



FR4 




Figure 44d 



WO 2005/056759 



54/61 



PCT/US2004/040694 



Figure 45. CDR grafted ICR62 VL 
Figure 45a 

DIQMTQSPSSLSASVGDRVTITCRASQNINNYLGWYQQKPGKAPKRLIYNTNNLQTGVPSR 
FSGSGSGTEFTLTISSLQPEDFATYYCLQHNSFPTFGAGTKLEIK 



Figure 45b 




Figure 45d 



WO 2005/056759 



55/61 



PCTAJS2004/040694 



Figure 46. CDR grafted ICR62 VH 
Figure 46a 

EVQLVQSGAEVKKPGATVKISCKVSGFTFTDYKMHWVQQAPGKGLEWMGLVNPNSGYTI 
YAEKFQGRVTITADTSTDTAYMELSSLRSEDTAVYYCATLSPGGYYVMDAWGQGTLVTVS 
S 

Figure 46b 




Figure 46c 

FR1 FR2 FR3 FR4 













IT" 1 








i-1 


■ 1... 








L. 

1 _ . 






T - 

'i *~ 




















ir " 




.1.1 
I 


y 


; i! 

...... — 1- w . j j- 




i 

» . .. 




} ; 

M 

V- * 






' j 






T 


: — ■■■ • i r 


— - 




i v ™ 
i 






4. _ . 








V > 










I: 












■ i 


r-r? 



Figure 46d 



WO 2005/056759 



56/61 



PCT/US2004/040694 



* 3* 

IS 5 




;■ ■ 

c - 



t>_o'a a|*> o'u u a*t» t3t> u^o a'o vaju 



" ** ? *J* ?i - • »j 



J * I 
■ i 

! : ! 

hi 



ii 



- aj > oo|» » • •'• «Voj» a «'o i_ 

<T» aVVa.'a EC * « ■ « 

^:d±::c±x&:::: 



SSBBSSSBSBBB 



o o • o « e o a • p 0,0 nj • ■ c 

OOOOOOOOOOOO o' o o o o op o 

»- »■ «-V a a *■ »- rv'*^ > »■'»■ ► 

* * *M «> * 4 * *> r r * »'* * 

a aVca a-m » ?« g» » j» » 



in 



•ofoo-boo 



■Tt.-i — — . — n — r- 

»bA- fey- a ^ »- a^»r».»i a a a-. 

*~ *>f >j»">[>'> »■> » >>"> > > > > > 

m m a a a ■ a a_a a|a ■'«'»'■ m mm a 
_«'»oo»oo"ooo«_o'»o,»o 00 

* •'•J? * • • • " * * •«•(••'• • 

" *-*» »;>*>■ a > > >"> >^>'>.j» >:> 
a ( a • • a a a a a a a n a a'a a a'a 
4 «. a *t+ - ^i**"!* - t* 

a mm »'»-•• . a^a » a ..'a- a'a a a'a 

OOOO0,0 0OOO , 0OO«0009« 

► -a7<- -Bi-tTrn<-IK- - - 

> * » = a » > a »_3 a > a a J 
a a o o O ft ft" • gt't » » »o e 0" 
.a* oio'o a a o'a a'a d a a'a o b o < 

i x ss » s s 33 1 sVsY* 11: 

!*!!..!5n!!5S3!i!3. 

L • » 

53555353«3«»S5532*5£ 
!!2!Vi«n525S3H!3: 



s - 



C 
O 

3 
O 

CO 

O 

O 
LU 
X 

X 

> 

CM 
<£> 

q: 
o 



si 

CD 



J- 



IT 
11 



B a 

E "- 

»' aT' 



S3 
P- 

* * 

51 a 

I: 



ii^iiintiiiii 

, ..J- ai.,aa ». a a . 




'I 



,V«l>lLVJaV.V.'.l l . 



1 a a a'a a aa a a'a ■ a'a^ 



•> ■ ■*»'» a »[»»■: 




r r , : : ! f ' ■ " 

iiaujjnnissHJS! 



CM 



3 

03 

o 
o 

Ui 
X 



CN 
CO 

a: 
o 



o 
N- 

£ 
3 

CD 



it 

.1 ,» 
> » 



mmmmmm 



' a • • «ii(hiiii 



rr 
n - 

1.; 
P 




»•»•••« aVWrT »••• a a a «V» « 




aja ^» a aja'a ••'•••*••• ^a «•'•• • 



'■MMMU a MMHtHM • 
>.t«,t.» * * ***** ********** 



-m~b. mU\. >V> .V.»»'» , 



• HtMllttlH,«l . • a ^IH 
» fV««,.' a a|aVa^a a ■ 
t »'»•»»». n'tA a • 




* aoaaa£aa»aA*^aa*a*l*Ja , **ai 

1 S5H2Vg«35M».5.I2;*S*! 



(«»«*. tr»Bs»xafcsssr&:;t> ri 

J 5 355S 553 S 5355 5 S S 5*5 i-S : 

....... ..... .... 

3S!S!3!S!H22!.a!S!?!!£l 



•: >i »; *» i; 

ill- »«-.«»•.« D-l 



WO 2005/056759 



57/61 



PCT/US2004/040694 



Figure 48. L3ICR62VL 
Figure 48a 

DIQMTQSPSSLSASVGDRVTITCRASQNINNYLNWYQQKPGKAPKRLIYNTNNLQTGIPSRF 
SGSGSGTDYTLTISSLQPEDFATYFCLQHNSFPTFGAGTKLEIK 

Figure 48b 




Figure 48c 

FR1 FR2 FR3 FR4 




Figure 48d 



WO 2005/056759 



58/61 



PCT/US2004/040694 



Figure 49. H9ICR62VH 
Figure 49a 

QVQLQQSGPGLVKPGASVKVSCKGSGFTFTDYKIHWVRQAPGKSLEWMGYFNPNSGYST 
YNEKFQGRVTITADKSTDTAYMELSSLRSEDTAVYYCTRLSPGGYYVMDAWGQGTLVTVS 
S 



FR1 



Figure 49b 

FR2 . FR3 



FR4 




Figure 49c 























I j _i :! •' ; i j 


i\- 


! 

"i 


Hi 




l ; 

, J . j, 




- 't 


; ; _m J 


"V*" 

! j: 












K 






-n .- • 


4i-r- 






- ' '1 !i » 




h- ■ 4- r 


i-t — i - — 




--t-i- 1 


, . . 

















Figure 49d 



WO 2005/056759 



59/61 



PCT/US2004/040694 



Figure 50. H10ICR62VH 
Figure 50a 

QVQLVQSGAEVKKPGASVKVSCKGSGFTFTDYKIHWVRQAPGKSLEWMGYFNPNSGYST 
YNEKFQGRVTITADKSTDTAYMELSSLRSEDTAVYYCTRLSPGGYYVMDAWGQGTLVTVS 

S 



Figure 50b 




Figure 50d 



15.14 Confidentiality. 

(a) No Limited Partner shall disclose to any Person any information related to the 
General Partner, the Partnership, any Principal, any Parallel Investment Vehicle, any Alternative 
Investment Vehicle, any Portfolio Company or proposed Portfolio Company or any of their 
respective Affiliates, in each case, that is not publicly available (or that is publicly available as a 
result of a disclosure by such Limited Partner or any director, employee, officer, agent, legal, 
financial or tax advisor of such Limited Partner in violation of this Section 04444 ); provided, 
however, that nothing contained herein shall prevent any Limited Partner from furnishing (i) any 

| re quir e d information that such Limited Partner is required to provide to any governmental 

regulatory agency, self-regulating body or in connection with any judicial, governmental or other 
| regulatory proceeding or as otherwise required by any applicable law , rule or regulation 

(provided that any disclosure that is either (x) not to a governmental regulatory agency or (y) not 

on a confidential basis, shall require prior written notice thereof to the General Partner) or (ii) 

any information, so long as such disclosure is for a bona fide business purpose relating to the 

such Limited Partners investment in the Partnership of such Limit e d Partner , to directors, 

officers, employees and legal, financial and tax advisors of such Limited Partner who are 

informed of the confidential nature of the information and who agree to be bound by the 

provisions of this Section 04444 or who are otherwise .bound by substantially similar obligations 

of confidentiality, and each Limited Partner agrees to b e boun d h e r e bv j [fn^po^^uciii 

feu^rgm^^ 1 
limitation of the foregoing, each Limited Partner acknowledges that notices and reports to such 
Limited Partner hereunder may contain material non-public information concerning, among 
other things, Portfolio Companies, and agrees not to use such information other than in 
connection with monitoring its investment in the Partnership and agrees, in that regard, not to 
trade in publicly traded Securities on the basis of any such information. Furthermore, the 
Partners hereby acknowledge that pursuant to § 17-305(f) of the Act, the rights of a Limited 
Partner to obtain information from the Partnership shall be limited to only those rights provided 
for in this Agreement, and that any other rights provided under § 17-305(a) of the Act shall not 
be available to the Limited Partners or applicable to the Partnership. 

(b) In order to preserve the confidentiality of certain information disseminated by the 
General Partner or the Partnership under this Agreement that a FOIA Limited Partner [that i s 
s ub ject' to FQIA o r any Li m i ted Partner that has "one or more equity own e rs that are s ubj e ct to 
FyQlA (any such Lim i t e d Partn e r, a " FOIA Li mite d Partner ; )j is entitled to receive pursuant to the 
provisions of this Agreement, including, without limitation, quarterly, annual and other reports 
(other than Schedule K-ls), information provided to the Advisory Committee and any 
information provided at meetings of the Limited Partners, the General Partner may (i) provide to 
such FOIA Limited Partner access to such information only on the Partnership's (or 
Management Company's) website in password protected, non-downloadable, non-printable 

| format, er-(ii) require such FOIA Limited Partner to return any copies of information provided to 
it by the General Partner or the Partnership (including any subsequent copies made by such 
Limited Partner) to the extent not prohibited by applicable law or regulation or (iii) lifefl . 

(c) Notwithstanding the provisions of Section 04444, the General Partner agrees that 
each Limited Partner that (x) is a private fund of funds (or other similar private collective 
investment vehicle) having reporting obligations to its investors and (y) has, prior to the date on 



( Commen t [M2]: Move lo definitions 



^omment j [M 3]: :'- add mutual " 



which such Limited Partner was admitted to the Partnership, notified the General Partner in 

| writing that it is {electing the benefits of, this Section 1.1 (c) 1 5 . 14 (c) may, in order to satisfy such 
reporting obligations, provide the following information to its investors (but only to the extent 
that such investors are informed of the confidential nature of the information and either agree to 

| be bound by the provisions of this Section Q44t4-4 or are otherwise bound by substantially similar 
obligations of confidentiality): (i) the name and address of the Partnership; (ii) the fact that such 
Limited Partner is a limited partner of the Partnership and the Partnership's general investment 
strategy; (iii) the identity of the Qeneral Partner and any Principal; (iv) the final closing date of 
the Partnership; (v) the amount of such Limited Partner's Commitment; (vi) the total amount of 
such Limited Partner's Capital Contributions; (vii) the total amount of distributions received by 
such Limited Partner from the Partnership; (viii) such Limited Partner's net internal rate of return 
with respect to the Partnership's performance as a whole as prepared by such Limited Partner; 

| [(ix) the name of any Portfolio Company, a description of the business of such Portfolio 
Company and information regarding the industry and geographic location of such Portfolio 

| Company; and (x) the cost of the Partnership's investment in a Portfolio [Compariyjl. With 
respect to any disclosure referred to in clauses (i) through (x) above, each Limited Partner shall 
indicate that such disclosure was not prepared, reviewed or approved, by the General Partner or 
the [Parthfeship]. 

(d) Each Limited Partner shall promptly notify the General Partner if at any time such 
Limited Partner is or becomes subject to Section 552(a) of Title 5 of the United States Code 
(commonly known as the "Freedom of Information Act") or any public disclosure law, rule or 
| regulation of any governmental body or non-governmental regulatory entity that could require 
similar or broader public disclosure of confidential information provided to such Limited Partner 
(collectively such laws, rules or regulations, " FOIA "). To the extent that any such Limited 
Partner receives a request for public disclosure of any confidential Partnership information 
| provided to it, such Limited Partner agrees that: (i) it shall use its reasonable best efforts to (*w) 
promptly notify the General Partner of such disclosure request and promptly provide the General 
Partner with a copy of such disclosure request or a detailed summary of the information being 
requested, (yx) inform the General Partner of the timing for responding to such disclosure 
request, (zy) consult with the General Partner regarding the response to such disclosure request; 
(hz) it sha ll us e comm e rcial l y rea s onabl e effort s to oppose and prevent the requested disclosure 
unless (A) such Limited Partner is advis e d determines in good faith upon advice of by-counsel 
that there exists no reasonable basis on which to oppose such disclosure or (B) such disclosure 
| relates solely to the information contained in clauses (i) through fcviuijof Section I . I Cc ) 1 5.1 1(c) 
(and does not include any information relating to individual Portfolio Companies and/or copies 
of this Agreement or related documents); and (iii) notwithstanding any other provision of this 
Agreement, the General Partner may, in order to prevent any such potential disclosure that the 
General Partner determines in good faith is [likejy.tp q'ccurj, withhold all or any part of the 
information otherwise to be provided to such Limited Partner; provided, however, that the 
General Partner shall not withhold any such information if such Limited Partner confirms in 
writing to the General Partner, based upon advice of counsel, that compliance with the 
procedures in Section 1 . 1(b) l 5.1 Kb) is legally sufficient to prevent such potential disclosure. 



Comment [M4]:;ShouId not that this 
t:'eliectibn;be'1riclud^'ih''sub'scnption : 



Comment [M5]: Expand list: 
management fees, date of investment, 
j public Hqmdity.eyents - 

Comment [ M 16]: Add assurance that 



Comment [M7]: Update if additional 
items added but exclude any portfolio 
company information if listed above 



• ^Comment [M8]: Any lower standard? 



(e) The obligations and undertakings of each Limited Partner under this Section 
0 15.11 shall be continuing and shall survive termination of the Partnership and this Agreement. 
Any restriction or obligation imposed on a Limited Partner pursuant to this Section 044rM may 



be waived by the General Partner in its discretion. Any such waiver or modification by the 
General Partner shall not constitute a breach of this Agreement or of any duty stated or implied 
in law or in equity to any Limited Partner, regardless of whether different agreements are 
reached with different Limited Partners. 



(f) The parties hereto agree that irreparable damage would occur if the provisions of 
this Section 04444 were |^^^^[.. It is accordingly agreed that the parties hereto shall be 
entitled to see k an injunction or injunctions to prevent breaches of this Section 04444 and to 
enforce specifically the terms and provisions hereof in any court of the U.S. or any state having 
jurisdiction, in addition to any other remedy to which they are entitled at law or in equity. 



(g) Add Tax Carve out 



; also* agree, not to raise as , a defen sc - 



^--- { Formatted: Bullets and Numberin g J 



(h) Add Delaware 305(b) boilerplate about ability to withhold 



EXHIBIT K 



Pipeline and Utility Easements Deed - by Owners to Mineral Owners and 

Mineral Lessee 



KJbCUKlJllNU Kbl^Ubo 1 bU Jt5 Y . 




Chicago Title Company 




WHEN RECORDED MAIL TO: 




Esperson/Grimm 




c/o Kronick Moskovitz 




Tiedemann & Girard 




1675 Chester Avenue, Suite 320 




Bakersfield, CA 93301 




Attention: Teri A. Bjorn, Of Counsel. 




The undersigned Grantors declare: 


Documentary transfer tax is: $ 



( ) computed on full value of property conveyed, or 
( ) computed on full value less value of liens and 
encumbrances remaining at time of sale. 



( ) Unincorporated area of County of Solano, or 

() City of Rio Vista. 

GRANT OF PIPELINE AND UTILITY EASEMENTS 

THIS GRANT OF PIPELINE AND UTILITY EASEMENTS (this "Easement Deed") is 
made and entered into as of , 200 (the "Effective Date") by and among: 

(1) THE GRIMM-RIO VISTA FAMILY LIMITED PARTNERSHIP, a 

California limited partnership; RICHARD W. ESPERSON, also known as RICHARD W. 
ESPERSON, JR., and IRENE S. ESPERSON, TRUSTEES OF THE RICHARD W. 
ESPERSON AND IRENE SUE ESPERSON FAMILY TRUST DATED OCTOBER 17, 
1991; MARK ESPERSON; GARY ESPERSON; STEPHEN ESPERSON; and KIMBERLY 
ESPERSON (collectively the "Surface Owners"), the fee owners of that certain real property 
legally described in Exhibit A-l (the "Property"). 



- 1 - 



(2) RIO VISTA HILLS HOLDING COMPANY, LLC, a Delaware limited 
liability company ("RVHHC"), who has an option to purchase that portion of the Property 
shown and identified on Exhibit B as the Sale Property (the "Sale Property"). The remainder of 
the Property shown and identified on Exhibit B , other than the Sale Property, is referred to as the 
"Retained Property/' Surface Owners and RVHHC are collectively sometimes referred to herein 
as the "Grantors." 

(3) NORMA JEAN GRIMM, also known as JEAN HARRIS GRIMM, AS 
TRUSTEE OF THE TRIMM FAMILY TRUST DATED OCTOBER 4, 1990 - 
SURVIVORS TRUST; RICHARD W. ESPERSON, JR.; JOAN ESPERSON WEDDELL; 
DAVID SANTOS; RICHARD SANTOS; STEPHEN ESPERSON; GARY ESPERSON; 
MARK ESPERSON; SUSAN A. BORGESEN, formerly SUSAN A. WOODWORTH; 
SANDRA J. DICKSON, formerly SANDRA GRIMM; SHARON E. HARRIS, formerly 
SHARON GRIMM; STEPHEN A. GRIMM, formerly STEPHEN GRIMM; THE GRIMM- 
RIO VISTA FAMILY LIMITED PARTNERSHIP, a California limited partnership; JENA 
HARRIS GRIMM; DAVID L. SANTOS, SURVIVING TRUSTEE OF THE DAVID L. 
AND LAURA E. SANTOS REVOCABLE TRUST DATED FEBRUARY 12, 2002; and 
RICHARD W. ESPERSON, also known as RICHARD W. ESPERSON, JR., and IRENE S. 
ESPERSON, TRUSTEES OF THE RICHARD W. ESPERSON AND IRENE SUE 
ESPERSON FAMILY TRUST DATED OCTOBER 17, 1991 (collectively the "Mineral 
Owners," each being a "Mineral Owner"), the owners of the reversionary interest in that certain 
mineral estate (the "Mineral Estate") in, on and under the Property and comprised of that portion 
of Parcel One and all of Parcels Two, Three and Four shown within bold line on the diagram 
attached as Exhibit C-l , which owners are listed as to each of such Parcels One, Two, Three and 
Four in Exhibit C-2 . The Mineral Estate is legally described in Exhibit C-3 . 

(4) The owner of the fee simple determinable interest in the Mineral Estate (by virtue 
of the grant of oil and gas lease concerning the Mineral Estate), ROSETTA RESOURCES 
OPERATING, L.P., a. Delaware limited partnership, successor to Calpine Natural Gas 
California, Inc., and Sheridan California Energy, Inc., both California corporations (the "Mineral 
Lessee"). For this Easement Deed, the exclusive fee simple determinable interest held by 
Mineral Lessee may be referred to as a mineral leasehold interest. The mineral leasehold estate 
of Mineral Lessee also includes rights in and to the real property located outside of the bold line 
on Exhibit B-l (the "Adjacent Property") and rights in and to that mineral estate in, on and under 
the Adjacent Property (the "Adjacent Mineral Estate"), which together with Mineral Lessee's 
leasehold rights in and to the Property and the Mineral Estate is referred to collectively as the 
"Leasehold Estate." The Leasehold Estate encumbers all of the Parcels shown on Exhibit C-l 
and is legally described in Exhibit C-4 . Mineral Owners and Mineral Lessee are collectively 
referred to herein sometimes as the "Grantees." 

Surface Owners, RVHHC, Mineral Owners and Mineral Lessee are sometimes 
collectively referred to herein as the "Parties" or individually as a "Party." 



RECITALS: 



A. Surface Owners own the Property, excepting the Mineral Estate, which is 
comprised of approximately 504 assessed acres of undeveloped real property in the City of Rio 
Vista ("City"), County of Solano, State of California, Assessor's Parcel Nos. 49-310-040, 49- 
3 1 0-300, 49-3 1 0-020 and 49-3 10-010. A diagram of the Property is attached as Exhibit B . 

B. Surface Owners have entered into an Option Agreement (the "Option Agreement") 
with RVHHC to sell the Sale Property for development of a residential/commercial project, 
including school sites, recreation/park sites, and open space areas and public uses (the "Project") as 
generally shown on Exhibit D . The Retained Property shown on Exhibit B will be retained for 
development by Surface Owners as" part of an overall master-planned community to be known as 
Del Rio Hills, which is currently planned to contain only the Sale Property and the Retained 
Property. This Easement Deed covers the Property which includes both the Sale Property and the 
Retained Property. 

C. As used in this Easement Deed, "Owner" means both Surface Owner and RVHHC 
and their respective heirs, assigns, transferees and successors. However, for purposes of 
apportioning responsibility and liability for the obligations generally ascribed to "Owner" in this 
Easement Deed, and unless the context of this Easement Deed otherwise specifically provides, (i) 
the fee estate owner of the Sale Property shall be responsible and liable only for those obligations, 
actions and activities arising under this Easement Deed that pertain to or otherwise stem from the 
ownership of the Sale Property and (ii) the fee estate owner of the Retained Property shall be 
responsible and liable only for those obligations, actions and activities arising under this Easement 
Deed that pertain to or otherwise stem from the ownership of the Retained Property. If the Option 
Agreement terminates or is terminated prior to RVHHC acquiring any portion of the Property, 
the term RVHHC shall mean Surface Owners or future developers taking their interest through 
Surface Owners, and RVHHC shall not have any further rights under the Accommodation 
Agreement (as defined in Recital G herein) or this Easement Deed. 

D. All oil, natural gas, casinghead gas, condensate and other hydrocarbon substances in, 
on and under the Property are part of the Mineral Estate. 

E. Mineral Lessee is the successor lessee under an Oil and Gas Lease between 
Edward Drouin, also known as E. D. Drouin, as lessor, and Amerada Petroleum Corporation of 
California, as lessee, dated May 28, 1935 (the "Lease"), and recorded on October 9, 1935, in 
Book J 51, Page 72, Official Records of Solano County, California. The Lease covers 912 acres, 
including the entire Property (504 acres) and the Adjacent Property (408 acres). The Leasehold 
Estate granted to Mineral Lessee by the Lease includes a fee simple determinable interest in the 
Mineral Estate, including the exclusive right to prospect, explore, test (including seismic and 
geologic investigations), drill for, produce, mine, extract, transport and remove oil, natural gas, 
casinghead gas, and other hydrocarbon substances from the Property and the Adjacent Property. 
Pursuant to the Lease, Mineral Lessee has the right of ingress and egress to and from the surface 
of the Property and the Adjacent Property as necessary (consistent with the terms and limitations 
of the Lease and applicable law) to conduct prospecting, exploration, testing (including seismic 
and geologic investigations), drilling for, producing, mining, extracting, transporting and 
removing oil, natural gas, casinghead gas, and other hydrocarbon substances in, on or under the 



Property and Adjacent Property. Said rights and activities are referred to herein as the 
"Extraction Operations." 

F. The Mineral Owners are the owners of the reversionary interest to the Mineral 
Lessee's interest in the Mineral Estate and are the successors to the lessor of the Lease. 

G. The Mineral Estate and the Leasehold Estate are subject to that certain Unit 
Agreement, Rio Vista Gas Unit, Contra Costa, Sacramento and Solano Counties, California, 
dated June 3, 1964, and recorded July 9, 1964, at Book 1280, Page 451, Official Records of 
Solano County (the "Unit Agreement"). Mineral Owners are parties to the Unit Agreement and 
receive a share of royalties for gas produced within the lands covered by the Unit Agreement, 
said lands defined as the "Unit Area" in the Unit Agreement. Mineral Lessee is the current Unit 
Operator, as defined in the Unit Agreement, and the current sole Working Interest Owner, as 
defined in the Unit Agreement. Mineral Lessee operates numerous existing natural gas wells in 
arid below the Unit Area, and has the right under the Unit Agreement to conduct Extraction 
Operations within the Unit Area. Mineral Owners and Mineral Lessee's predecessors-in-interest 
entered into the Unit Agreement. Mineral Lessee has rights under the Unit Agreement, and 
various other leases, to explore, develop and extract natural gas, condensate and associated 
hydrocarbons within the Unitized Formation of the Unit Area as defined in the Unit Agreement, 
as well as the right of ingress and egress to and from the surface of the Unit Area as provided in 
the Unit Agreement and relevant leasehold agreements. 

H. As of the Effective Date, Mineral Lessee has fourteen (14) existing gas well sites 
on the Property (each containing a well) and plans for six (6) additional well sites on the 
Property (these twenty (20) well sites are referred to collectively as the "Well Sites"). A Well 
Sites Plan shows the locations of all twenty (20) Well Sites, and is attached as Exhibit E . For 
purposes of this Easement Deed, each of the Well Sites is a "Well Site." The Well Sites will be 
used to support existing wells, and to drill new wells. Pipelines presently exist on the Property to 
support Mineral Lessee's existing Well Sites, and Mineral Lessee needs additional new pipelines 
on the Property to support the Well Sites (the pipelines presently existing on the Property and 
new pipelines planned by Mineral Lessee on the Property are referred to collectively as the 
"Pipelines"). Pursuant to the Lease and the Unit Agreement, Mineral Lessee also has numerous 
existing gas wells and pipelines located on the Adjacent Property, and has plans for additional 
wells and pipelines on the Adjacent Property. 

L Mineral Lessee is required to obtain certain approvals and/or permits ("City 
Extraction Approvals") from the City for the development of new natural gas facilities, including 
new wells and redrilling and deepening existing wells, pursuant to Chapter 13.12 of the City's 
Municipal Code ("Natural Gas Ordinance" or "NGO"). Mineral Lessee may also need to obtain 
approvals from the California Department of Conservation, Division of Oil, Gas and Geothermal 
Resources and possibly from other agencies such as the U.S. Fish and Wildlife Service and the 
California Department of Fish and Game for these drilling activities within the Well Sites 
("Other Agency Extraction Approvals"). As used in this Easement Deed, the terms City 
Extraction Approvals and Other Agency Extraction Approvals shall concern only such drilling 
activities within the Property (and not drilling activities within the Adjacent Property for which 
Mineral Lessee is required to seek separate approvals). 



J. In conjunction with the development of the surface of the Property for activities, 
structures and uses that are not related to mineral extraction ("Surface Development"), Owners 
are required to obtain approvals and/or permits from the City (including a subdivision map, 
pursuant to California Government Code Section 66410 et seq.), and possibly other 
governmental agencies, for Surface Development and related activities, construction and 
operations (collectively "Surface Development Approvals"). 

K. As a general matter, and subject to the terms and limitations of the Lease and 
applicable law, Mineral Lessee currently has non-exclusive rights to use the surface of the 
Property for the purposes set forth in the Lease and the Unit Agreement. Absent an agreement to 
specify which portion(s) of the Property can be used by which Party for what purpose(s), a 
possibility exists for conflict between and among the Parties as each exercises its rights in the 
Property. Accordingly, Mineral Owners and Mineral Lessee desired assurances and certainty 
that they will be able to continue existing Extraction Operations on certain portions of the 
Property, and in and below the Unit Area, under the terms and conditions of the Lease and the 
Unit Agreement and will be able to develop new Extraction Operations within the Well Sites, 
and in and below the Unit Area, under the terms and conditions of the Lease and the Unit 
Agreement. Similarly, Owners desire assurances and certainty that the Extraction Operations on 
the Property will be confined to certain areas of the Property, so that Owners can proceed with 
Surface Development. 

L. The Parties 5 desired assurances and certainties are set forth in that certain 
Accommodation Agreement - Del Rio Hills ("Accommodation Agreement"), recorded on 
at Series # in the Official Records of Solano County. 

M. Among other matters, the Accommodation Agreement (at Section 6.h) granted 
Mineral Owners and Mineral Lessee a temporary, unrestricted easement (the "Temporary Floating 
Pipeline/Utility Easement") over all of the Property for purposes of installing, operating and 
maintaining pipelines and related appurtenances necessary and convenient for Mineral Lessee's 
operations within the Property, the Unit Area and elsewhere. The Temporary Floating 
Pipeline/Utility Easement arose automatically and without the need for further documentation or 
approval upon the Parties' execution of the Accommodation Agreement; the Parties covenanted 
and agreed that the recordation of the Accommodation Agreement constituted sufficient and 
appropriate evidence of the grant of the Temporary Floating Pipeline/Utility Easement. The 
Temporary Floating Pipeline/Utility Easement will continue in full force and effect until such 
time as one or more final subdivision maps are recorded for Surface Development. Upon 
recordation of a final subdivision map for Surface Development, the Temporary Floating 
Pipeline/Utility Easement for only the portion of the Property which is the subject of the final 
subdivision map will automatically terminate and be of no further force or effect. The 
Temporary Floating Pipeline/Utility Easement for those portions of the Property which are not 
the subject of a final subdivision map for Surface Development will continue in full force and 
effect. Notwithstanding said automatic termination, Mineral Lessee and Mineral Owners will 
execute and deliver an appropriate instrument or document manifesting release of all or portions, 
as applicable, of the Temporary Floating Pipeline/Utility Easement following Owner's delivery 



-5- 



of (i) a written request for such instrument or document, and (ii) a copy of the recorded final 
subdivision map for Surface Development of the Property. 

N. Pursuant to the Accommodation Agreement, Mineral Owners and Mineral Lessee 
desire to obtain exclusive easements across certain portions of the Property in order to establish, 
develop, maintain and repair and access Well Sites and to conduct Extraction Operations. 

AGREEMENT : 

In consideration of the mutual promises contained in this Easement Deed and of other 
valuable consideration, the receipt and sufficiency of which the Parties expressly acknowledge, it is 
agreed as follows: 

1. Grant of Pipeline and Utility Easements . 

Grantors hereby gratnt to Grantees temporary and permanent nonexclusive 
easements that are reasonably necessary for: (i) all pipelines and utilities necessary to service 
each Well Site, including without limitation, installation of conduits, pipe in pipe and wire in 
pipe; (ii) all Well Site flow lines and utility lines inside of protective conduit; (iii) pipeline stub- 
outs and utility or pipeline connections; and (iv) ingress and egress over and under the easement 
area for the installation, construction, operation, repair, maintenance, relocation and replacement 
of all pipelines and utilities (the "Pipeline/Utility Easements"). The Pipeline/Utility Easements 
shall be in gross and shall benefit Mineral Lessee and Mineral Owners, and are hereby conveyed 
to Mineral Owners and Mineral Lessee for non-exclusive use by Mineral Owners and Mineral 
Lessee. The Pipeline/Utility Easements as shown on the Well Sites Plan shows the Parties' 
agreed upon location of temporary and permanent easements which shall consist of (i) a 
permanent easement area a minimum of twenty (20) feet wide; and (ii) a temporary construction 
easement area to be used by Mineral Lessee during construction of pipelines and access 
roadways of an additional fifteen (15) feet on both sides of the twenty (20) foot permanent 
easement. The Pipeline/Utility Easements are designed to ensure, and shall have a scope 
sufficient to ensure, that any gas produced from any location (whether or not from below the 
Property, the Unit Area or elsewhere) that flows into the pipeline system on the Property has at 
least two outlets to exit the system on the Property. 

2. Pipeline and Utility Construction . 

a. If Mineral Lessee Constructs its Improvements in an Area Before Owners 
Construct their Improvements in the Same Area . 

At all times, but prior to Surface Development in the vicinity of the area of the 
Property where Mineral Lessee desires to construct and install a pipeline or other utilities, 
Mineral Lessee shall be responsible for the installation and cost of all improvements necessary to 
transport production from its wells and to support its Well Sites with utility service. Mineral 
Lessee shall construct such improvements in the location of the Pipeline/Utility Easements set 
forth on the Well Sites Plan. Owners shall be entirely responsible to ensure that Mineral 
Lessee's improvements are not disturbed by any subsequent improvements, including grading, of 
the surface by Owners. In the event that Owners and Mineral Lessee agree, after Mineral Lessee 



-6- 



has installed pipelines and utilities, to modify the locations of any Pipeline/Utility Easements to 
accommodate Owners' Surface Development, then Owners shall be responsible for all costs of 
any resulting relocation of the pipelines and utilities. Mineral Lessee shall have the right to 
approve the time and manner of any relocation of pipelines or utilities done by Owners, or at 
Owners' request, prior to such relocation; such approval shall not be unreasonably withheld, 
conditioned or delayed, provided such relocation will not result in any substantial or material 
interruption of pipeline or utility service to Mineral Lessee's Extraction Operations on the 
Property. For purposes of this Easement Deed, "substantial or material interruption" shall mean 
a complete shut down, for a period exceeding forty-eight (48) consecutive hours, of one or more 
of Mineral Lessee's actively producing gas wells which utilized the affected Pipeline/Utility 
Easements, in spite of Mineral Lessee's commercially reasonable efforts to re-route gas 
production from the affected gas well(s) using other existing pipelines. Mineral Lessee shall 
issue its approval or disapproval within thirty (30) days of receipt of Owner's plans and 
specifications for the relocation of any pipelines or utilities. Mineral Lessee acknowledges and 
agrees that there will likely be temporary disruption of pipeline or utility service to the 
Extraction Operations on the Property when the new pipelines and utility connections are made. 
In the event that any such relocation activities result in a substantial or material interruption, 
Owner shall pay to Mineral Lessee a fee equal to Mineral Lessee's lost production value from 
the affected gas well(s) for the period (i) beginning at the expiration of the initial forty-eight (48) 
consecutive hours, and (ii) ending when the pipeline connection is fully restored to the actively 
producing gas well(s). For purposes of this Section 2. a, the fee payable to Mineral Lessee for the 
lost production value of an affected gas well shall be determined by the average of the affected 
gas well's daily yield for the thirty (30) day period preceding the date of the disruption, and 
based upon the contract price Mineral Lessee is then receiving from the purchaser of the gas 
well's production. 

b. If Owners Construct their Improvements in an Area before Mineral Lessee 
Constructs its Improvements in the Same Area . 

At all times after Owners have commenced development of the Property 
consistent with the Accommodation Agreement and the Surface Development Approvals on or 
near any of Mineral Lessee's Pipeline/Utility Easements, Owners shall install improvements 
(such as pipelines, water lines, telemetry/telephone lines and electrical lines) within the 
Pipeline/Utility Easements according to the Well Sites Plan or Survey, as appropriate (see ) 
Section 1 .f of the Accommodation Agreement), to service existing or future wells at the Property 
as shown on the Well Sites Plan or Survey, as appropriate. Owners shall be responsible for the 
construction costs and installation of such pipelines and utilities; Mineral Lessee shall be 
responsible for the materials costs of the pipeline and other utility materials. The intent of this 
provision is to allow efficiencies of constructing Mineral Lessee's pipelines and other 
underground utilities at the same time Owners are building streets, sidewalks, and other 
infrastructure improvements. Prior to construction, Owners shall submit plans for such pipelines 
and other utility improvements to Mineral Lessee for Mineral Lessee's review and approval to 
ensure that the pipelines and improvements meet or comply with Mineral Lessee's specifications 
and standards and/or requirements of any approval or permit granted to Mineral Lessee for 
Extraction Operations. Mineral Lessee shall not unreasonably withhold, condition or delay 
approval of Owners' plans for such pipelines and other utility improvements, and Mineral Lessee 
shall issue its written approval or disapproval within thirty (30) days of receipt of Owners' plans 



-7- 



and specifications. If Mineral Lessee disapproves of the proposed plans, Mineral Lessee's 
written notice to Owners shall specify the nature of Mineral Lessee's objections and identify 
proposed revisions to the plans to correct the objections. Within thirty (30) days following 
receipt of Mineral Lessee's disapproval notice and proposed revisions, Owners shall notify 
Mineral Lessee in writing concerning whether Owners accept or reject the proposed revisions. If 
Owners accept the proposed revisions, the plans shall be duly modified and implementation of 
the pipeline improvements shall proceed accordingly. If Owners reject the proposed revisions, 
Owners and Mineral Lessee shall resolve their outstanding issues in accordance with the dispute 
resolution provisions set forth in Sections 12.a and 12.b of the Accommodation Agreement. 
[This reference to the AA may work, as it relates to the construction phase, when A A still on title, 
prior to any conveyances of the areas burdened by the easements (open space, roads, etc.) to the 
City.] 

3. Pipeline and Utility Relocation . 

a. Pipeline Location . Prior to the recordation of a final subdivision map for 
Surface Development, Mineral Lessee shall have the right to locate pipelines and utilities under 
the Temporary Floating Pipeline/Utility Easement, as granted by the Accommodation Agreement 
and described in Recital M of this Easement Deed. The final location of the Pipeline/Utility 
Easements shall be determined by those certain City Extraction Approvals, a development 
agreement (with a term of at least fifteen (15) years) between Mineral Lessee and the City, 
pursuant to California Government Code Section 65864 et seq., those Other Agency Extraction 
Approvals, and the development agreement with the City vesting those extraction approvals 
("Development Agreement"), with potential modification as set forth herein to accommodate 
Owners' Surface Development. 

b. Relocation of Existing Pipelines . The location of some of the 
Pipeline/Utility Easements may need to change from time to time as Owners obtain approvals 
and/or permits from the City and other governmental agencies for Surface Development over 
time. Owners may, at any time, request that certain existing (as of the Effective Date) utilities 
and pipelines located within the Property ("Existing Utilities") that conflict with any of Owners' 
Surface Development Approvals be abandoned and, as appropriate, removed by Owners. These 
utilities and pipelines may be owned by Mineral Lessee or by other parties such as Pacific Gas 
and Electric Company ("PG&E") (utilities and pipelines owned by PT&E or other third parties 
are referred to herein as "Third Party Utilities"); Mineral Lessee may have the right to use Third 
Party Utilities. Prior to abandonment and removal of any Existing Utilities, Owners must obtain 
the written consent of the owner of the Existing Utility and, in the instance of Third Party 
Utilities, of Mineral Lessee. If Mineral Lessee, and, if applicable, the owner of a Third Party 
Utility, agree to the abandonment and removal, then Mineral Lessee shall not be responsible for 
the abandonment and removal or any costs of same, and such costs shall be paid by Owners 
and/or, if applicable, the owner of a Third Party Utility, as agreed between those parties. In the 
event any of Mineral Lessee's pipelines or utilities need to be relocated and abandoned, such 
pipeline shall not be abandoned until a replacement pipeline or utility of sufficient size, quality 
and location has been installed and commissioned (if deemed necessary by Mineral Lessee) to 
transport gas produced from any location (whether or not from below the Property, the Unit Area 
or elsewhere) that flows into the pipeline system on the Property. 



-8- 



c. Relocation of Pipelines or Utilities After Completion of Surface 
Development , At all times after Owners have completed Surface Development of the Property, 
any pipelines or utility improvements that Mineral Lessee needs to replace, repair or improve 
(presuming such need was not caused by Owners, in which case Owners shall be entirely 
responsible for the costs to replace, repair or improve) shall be done at Mineral Lessee's sole 
cost. Where there are surface improvements in the area of the Pipeline/Utility Easements, 
including without limitation, landscaping, Mineral Lessee shall be responsible for all incremental 
costs associated with construction conflicts caused by such improvements and Mineral Lessee 
shall be responsible for restoring the surface to a condition comparable to that which existed 
prior to Mineral Lessee's construction activities and for replacing any surface improvements 
damaged by Mineral Lessee's activities. 

4. Environmental Indemnification . 

With regard to their respective activities on the Property as permitted by this 
Easement Deed, the Parties shall comply with all environmental laws, statutes and regulations 
issued by federal, state and local governmental agencies ("Agencies") and shall conduct their 
activities on the Property according to environmental standards set by the Agencies, including 
without limitation, hazardous waste, toxic substances, hydrocarbons and petroleum based 
substances, water, wetlands, endangered and threatened species and air standards ("Agency 
Environmental Standards"). The Parties Lessee shall also comply with California Health and 
Safety Code Section 25359.7. If a Party's use of or activity on the Property results in a loss to 
any other Party due to a violation of Agency Environmental Standards resulting from use of the 
Property, whether an enforcement action is brought by an agency or a private citizen, the Party 
whose use caused the loss shall defend (using legal counsel reasonably satisfactory to the other 
Party), indemnify and hold harmless the other Party(ies) from the loss and any resulting penalties 
and fines; and the Party whose use led to the violation shall, at its sole cost, respond to all such 
violations for its benefit and for the benefit of the other Party(ies), including all clean-up, 
demolition, detoxification, and disposal and the preparation of any closure or other required 
plans. In the case of joint or contributory negligence, breach of contract or other fault or strict 
liability on the part of the Party seeking indemnification, principles of comparative negligence 
shall be followed and each Party shall bear the proportionate cost of any loss, damage, expense, 
or liability attributable to such Party's negligence, breach of contract, use or other fault or strict 
liability. This Section 4 applies only to activities and uses occurring after the Effective Date. 
There are no third party beneficiaries to this Section 4. The environmental indemnification 
obligations contained in this Section 4 shall survive the termination of each Parties' interest in 
the Property and the Pipeline/Utility Easements. 

5. Termination of the Pipeline/Utility Easements . 

a. Mineral Lessee's interest in the Pipeline/Utility Easements shall terminate 
when Mineral Lessee releases the Pipeline/Utility Easements to Owners and Mineral Owners. 
Mineral Lessee shall have the right, in its sole discretion, to elect to abandon any pipeline in 
place. Upon expiration or termination of its interest in the Pipeline/Utility Easements, Mineral 
Lessee or its successor, shall promptly and expeditiously execute and acknowledge, on request of 
Owners, Mineral Owners or their title insurer, a release to Mineral Owners formally releasing of 
record Mineral Lessee's interests in, and rights under, the Pipeline/Utility Easements. 



-9- 



b. Mineral Owners' interest in the Pipeline/Utility Easements shall terminate 
at the earlier of (i) when the pipelines are not used to transport hydrocarbons for a period of 
longer than one hundred eighty (180) days after Mineral Lessee has recorded a release of the 
Pipeline/Utility Easements, (ii) when the ownership of the Mineral Estate and Surface Estate 
merge, or (iii) when the Mineral Owners' interest in the Pipeline/Utility Easements is otherwise 
released. 

6. Binding Effect , 

The provisions of this Easement Deed shall be binding upon and inure to the 
benefit of the Parties, their respective heirs, transferees and successors in interest, including, 
without limitation, all subsequent owners of the Sale Property and the Retained Property, and the 
burden of the Pipeline/Utilities Easements shall be covenants running with the land of the 
Property. For purposes of apportioning responsibility and liability for the obligations generally 
ascribed to "Owner" in this Easement Deed, and unless the context of this Easement Deed 
otherwise specifically provides, (i) the fee estate owner of the Sale Property shall be responsible 
and liable only for those obligations, actions and activities arising under this Easement Deed that 
pertain to or otherwise stem from the ownership of the Sale Property; and (ii) the fee estate 
owner of the Retained Property shall be responsible and liable only for those obligations, actions 
and activities arising under this Easement Deed that pertain to or otherwise stem from the 
ownership of the Retained Property. If the Option Agreement terminates or is terminated prior to 
RVHHC acquiring any portion of the Property, the term RVHHC shall mean Surface Owners or 
future developers taking their interest through Surface Owners, and RVHHC shall not have any 
further rights under this Easement Deed. Mineral Lessee shall have the right to assign this 
Easement Deed and any of Mineral Lessee's rights hereunder. 

7. Mineral Lessee's Right to Authorize Use by Unit Operator; Mineral Lessee and 
Mineral Owners' Right to Authorize Use by Third Party Utility Providers . 

Mineral Lessee shall have the right to authorize the use of the Pipeline/Utility 
Easements by the Unit Operator for the purposes of conducting Unit operations, so long as the 
Unit Agreement is in effect. Mineral Lessee and Mineral Owner shall have the right to authorize 
the use of the Pipeline/Utility Easements by any public or private utility company providing any 
utilities serving the Well Sites, or as necessary to transport gas to market. 

7. Status of Title . 

This Easement Deed is made subject to all conditions, covenants, restrictions, 
leases, easements, licenses, liens, encumbrances and claims of title of record which may affect 
the Pipeline/Utility Easements. 

8. Notices . 

All notices, demands, or other communications required or permitted by this 
Easement Deed or by law to be served on or given to a Party shall be in writing and shall be 
deemed delivered (i) if personally delivered, upon delivery to the Party to whom directed, (ii) if 
mailed, upon the expiration of forty-eight (48) hours from the date of mailing in the United 



- 10- 



States mail, registered or certified, return receipt requested, addressed to the Party address shown 
below, or (iii) if faxed, upon receipt of the fax transmittal, transmitted to the Party fax number 
shown below, provided the machine sending the fax provides a written confirmation of receipt: 



If to the Surface Owners, at: 



And to: 



And to: 



With a copy to: 



And with a copy to: 



If to Mineral Owners, at: 



And to: 



And to: 



c/o Mr. John Wyro 
The Wyro Company 
40 Valley Drive 
Orinda,CA 94563 
Telephone: 925-254-5246 
Fax: 925-254-5299 

Richard and Sue Esperson 
398 Crescent Drive 
Rio Vista, CA 94571 

Mrs. Jean Grimm 
35 San Gabriel Dr. 
Fairfax, CA 94930 

Teri A. Bjorn, Of Counsel 

Kronick, Moskovitz, Tiedemann & Girard 

1675 Chester Avenue, Suite 320 

Bakersfield, CA 93301 

Telephone: 661-864-3800 

Fax: 661-864-3810 

David G. Kenyon, Esq. 
7200 Redwood Blvd., Suite 404 
Novato,CA 94945 
Telephone: 415-892-1868 
Fax: 415-892-1716 

c/o Mr. John Wyro 
The Wyro Company 
40 Valley Drive 
Orinda,CA 94563 
Telephone: 925-254-5246 
Fax; 925-254-5299 

Richard and Sue Esperson 
398 Crescent Drive 
Rio Vista, CA 94571 

Mrs. Jean Grimm 
35 San Gabriel Dr. 
Fairfax, CA 94930 



- 11 - 



With a copy to: 



And with a copy to: . 



If to Mineral Lessee, at: 



With a copy to: 



And with a copy to: 



If to the RVHHC, at: 



And to: 



teri A. Bjorn, of Counsel 

Kronick, Moskovitz, Tiedemann & Girard 

1675 Chester Avenue, Suite 320 

Bakersfield, CA 93301 

Telephone: 661-864-3800 

Fax: 661-864-3810 

David G. Kenyon, Esq. 
7200 Redwood Blvd., Suite 404 
Novato, CA 94945 
Telephone: 415-892-1868 
Fax: 415-892-1716 

Rosetta Resources Operating, L.P. 
1200 17th Street, Suite 770 
Denver, CO 80202 
Telephone: (720) 946-1315 
Fax:(720)359-9140 

Rosetta Resources, Inc. 
Attn: General Counsel 
717 Texas Avenue, Suite 2800 
Houston, TX 77002 
Telephone: (713) 335-4017 
Fax: (7 13) 335-4136 

Stephen R. Finn, Esq. 
One Market Street, Spear Tower 
San Francisco, CA 94105 
Telephone: 415-442-1251 
Fax: 415-442-1001 

Rio Vista Hills Holding Company, LLC 
c/o Lewis Operating Corp. 
9216Kiefer Blvd, Suite 8 
Sacramento, CA 95827 
Attn: Douglas Mull, Vice Pres. 
Telephone: 916-363-2617 ext.226 
Fax: 916-364-9353 

Lewis Operating Corp. 

Legal Department 

1 1 56 N. Mountain Avenue 

Upland, CA 91786 

Attn: W. Bradford Francke, Esq. 



- 12- 



Telephone: 909-946-7538 
Fax: 909-946-6725 



With a copy to: Law Offices of Gregory D. Thatch 

1730 T* Street, Suite 220 
Sacramento, C A 95814 
Attn: Michael Devereaux, Esq. 
Telephone: 916-443-6956. 
Fax: 916-443-4632 



9. Choice of Law; Venue . 

This Easement Deed shall be governed by and construed in accordance with the 
substantive and procedural laws of the State of California, excluding any laws that require the 
application of another jurisdiction's laws. This Easement Deed is entered into and is to be 
performed in Solano County, California, arid accordingly the only appropriate venue for a 
dispute under this Easement Deed is in the Solano County Superior Court of California. All 
references to codes in this Easement Deed are to California or City codes, unless otherwise 
specified. 

10. Entire Agreement . 

With the exception of the Lease, Unit Agreement, Accommodation Agreement 
and Declaration of Covenants, Conditions and Restrictions for the Del Rio Hills Project, this 
Easement Deed (including the Exhibits to this Easement Deed) supersedes any and all other 
agreements, either oral or in writing, between the parties with respect to the subject matter and 
contains all of the covenants and agreements between the parties with respect to such matter, and 
each party to this Easement Deed acknowledges that no representations, inducements, promises 
or agreements, oral or otherwise, have been made by any party, or anyone acting on behalf of 
any party, which are not embodied herein, and that no other agreement, statement or promise not 
contained in, or contemplated by this Easement Deed shall be valid or binding. 

11. Modification . 

This Easement Deed may be modified or amended only by a written recordable 
document signed and acknowledged by the (a) then-current Surface Owners, (b) RVHHC or its 
successor, if any, (c) then-current Mineral Lessee, and (d) then-current Mineral Owners, or by their 
respective successors-in-interest, transferees or assigns. 

12. Partial Invalidity . 

In the event a court of law determines that any provision of this Easement Deed, or 
portion thereof, is prohibited, unlawful, or unenforceable under any applicable law of any 
jurisdiction, the remainder of the provisions hereof shall remain in full force and effect and shall 
in no way be affected, impaired or invalidated. 



- 13- 



13. Time . 

Time is of the essence in the performance of the parties* respective obligations 
contained in this Easement Deed. 

14. Counterparts . 

This Easement Deed may be executed in any number of counterparts all of which 
when executed shall constitute one document. 

IN WITNESS WHEREOF, Grantors and Grantees have executed this Easement Deed as of 
the Effective Date. 



GRANTORS: 

THE GRIMM-RIO VISTA FAMILY 
LIMITED PARTNERSHIP 

By_ 

General Partner 



- 14- 



RICHARD W. ESPERSON also known as 
Richard W. Esperson, Jr., as Trustee of the 
Richard W. Esperson and Irene Sue 
Esperson Family Trust dated October 17, 
1991 



IRENE S. ESPERSON, as Trustee of the 
Richard W. Esperson and Irene Sue 
Esperson Family Trust dated October 17, 
1991 



MARK ESPERSON 



GARY ESPERSON 



STEPHEN ESPERSON 



KIMBERLY ESPERSON 



RIO VISTA HILLS HOLDING 
COMPANY, LLC 

a Delaware limited liability company 

By North Mountain Corporation, 
a California corporation 
Its Sole Manager 

By 



By. 



GRANTEES: 



NORMA JEAN GRIMM, also known as 
Jean Harris Grimm, as Trustee of the Grimm 
Family Trust dated October 4, 1990 - 
Survivors Trust 



RICHARD W. ESPERSON, JR. 



JOAN ESPERSON WEDDELL 



DAVID SANTOS 



RICHARD SANTOS 



STEPHEN ESPERSON 



GARY ESPERSON 



MARK ESPERSON 



SUSAN A. BORGESEN, formerly Susan A. 
Woodworth 



SANDRA J. DICKSON, formerly Sandra 
Grimm 



SHARON E. HARRIS, formerly Sharon 
Grimm 



STEPHEN A. GRIMM, formerly Stephen 
Grimm 



THE GRIMM-RIO VISTA FAMILY 
LIMITED PARTNERSHIP 

By 

General Partner 



JEAN HARRIS GRIMM 



DAVID L. SANTOS, as Surviving Trustee 
of the David L. and Laura E. Santos 
Revocable Trust dated February 12, 2002 



RICHARD W. ESPERSON, also known as 
Richard W. Esperson, Jr., as Trustee of the 
Richard W. Esperson and Irene Sue 
Esperson Family Trust dated October 17, 
1991 



IRENE S. ESPERSON, as Trustee of the 
Richard W. Esperson and Irene Sue 
Esperson Family Trust dated October 17, 
1991 



ROSETTA RESOURCES OPERATING, 
L.P., a California limited partnership 

By : 



By.